1
|
Chen Y, Douanne N, Wu T, Kaur I, Tsering T, Erzingatzian A, Nadeau A, Juncker D, Nerguizian V, Burnier JV. Leveraging nature's nanocarriers: Translating insights from extracellular vesicles to biomimetic synthetic vesicles for biomedical applications. SCIENCE ADVANCES 2025; 11:eads5249. [PMID: 40009680 PMCID: PMC11864201 DOI: 10.1126/sciadv.ads5249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Naturally occurring extracellular vesicles (EVs) and synthetic nanoparticles like liposomes have revolutionized precision diagnostics and medicine. EVs excel in biocompatibility and cell targeting, while liposomes offer enhanced drug loading capacity and scalability. The clinical translation of EVs is hindered by challenges including low yield and heterogeneity, whereas liposomes face rapid immune clearance and limited targeting efficiency. To bridge these gaps, biomimetic synthetic vesicles (SVs) have emerged as innovative platforms, combining the advantageous properties of EVs and liposomes. This review emphasizes critical aspects of EV biology, such as mechanisms of EV-cell interaction and source-dependent functionalities in targeting, immune modulation, and tissue regeneration, informing biomimetic SV engineering. We reviewed a broad array of biomimetic SVs, with a focus on lipid bilayered vesicles functionalized with proteins. These include cell-derived nanovesicles, protein-functionalized liposomes, and hybrid vesicles. By addressing current challenges and highlighting opportunities, this review aims to advance biomimetic SVs for transformative biomedical applications.
Collapse
Affiliation(s)
- Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Tad Wu
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Ishman Kaur
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- École de technologie supérieure ÉTS, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - David Juncker
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | | | - Julia V. Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
2
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [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: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
3
|
Baudo G, Flinn H, Holcomb M, Tiwari A, Soriano S, Taraballi F, Godin B, Zinger A, Villapol S. Sex-dependent improvement in traumatic brain injury outcomes after liposomal delivery of dexamethasone in mice. Bioeng Transl Med 2024; 9:e10647. [PMID: 39036088 PMCID: PMC11256133 DOI: 10.1002/btm2.10647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 07/23/2024] Open
Abstract
Traumatic brain injury (TBI) can have long-lasting physical, emotional, and cognitive consequences due to the neurodegeneration caused by its robust inflammatory response. Despite advances in rehabilitation care, effective neuroprotective treatments for TBI patients are lacking. Furthermore, current drug delivery methods for TBI treatment are inefficient in targeting inflamed brain areas. To address this issue, we have developed a liposomal nanocarrier (Lipo) encapsulating dexamethasone (Dex), an agonist for the glucocorticoid receptor utilized to alleviate inflammation and swelling in various conditions. In vitro studies show that Lipo-Dex were well tolerated in human and murine neural cells. Lipo-Dex showed significant suppression of inflammatory cytokines, IL-6 and TNF-α, release after induction of neural inflammation with lipopolysaccharide. Further, the Lipo-Dex were administered to young adult male and female C57BL/6 mice immediately after controlled cortical impact injury (a TBI model). Our findings demonstrate that Lipo-Dex can selectively target the injured brain, thereby reducing lesion volume, cell death, astrogliosis, the release of pro-inflammatory cytokines, and microglial activation compared to Lipo-treated mice in a sex-dependent manner, showing a major impact only in male mice. This highlights the importance of considering sex as a crucial variable in developing and evaluating new nano-therapies for brain injury. These results suggest that Lipo-Dex administration may effectively treat acute TBI.
Collapse
Affiliation(s)
- Gherardo Baudo
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Hannah Flinn
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Morgan Holcomb
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Anjana Tiwari
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
| | - Sirena Soriano
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
| | - Francesca Taraballi
- Department of Orthopedics and Sports Medicine and Center for Musculoskeletal RegenerationHouston Methodist HospitalHoustonTexasUSA
| | - Biana Godin
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyHouston Methodist Research InstituteHoustonTexasUSA
- Department of Obstetrics and GynecologyWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTexasUSA
| | - Assaf Zinger
- Department of Cardiovascular SciencesHouston Methodist Research InstituteHoustonTexasUSA
- Department of Chemical EngineeringTechnion−Israel Institute of TechnologyHaifaIsrael
| | - Sonia Villapol
- Department of Neurosurgery and Center for NeuroregenerationHouston Methodist Research InstituteHoustonTexasUSA
- Department of Neuroscience in Neurological SurgeryWeill Cornell Medicine College (WCMC)New YorkNew YorkUSA
| |
Collapse
|
4
|
Wang B, Shen J, Wang X, Hou R. Biomimetic nanoparticles for effective Celastrol delivery to targeted treatment of rheumatoid arthritis through the ROS-NF-κB inflammasome axis. Int Immunopharmacol 2024; 131:111822. [PMID: 38503010 DOI: 10.1016/j.intimp.2024.111822] [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: 01/20/2024] [Revised: 02/19/2024] [Accepted: 03/05/2024] [Indexed: 03/21/2024]
Abstract
Previous study has indicated that Celastrol (Cel) has various physiological and pharmacological effects, including antibacterial, antioxidant, pro-apoptotic, anticancer and anti-rheumatoid arthritis (RA) effects. However, low water solubility, low oral bioavailability, narrow treatment window, and high incidence of systemic adverse reactions still limit the further clinical application of Cel. Here, aiming at effectively overcome those shortcomings of Cel to boost its beneficial effects for treating RA, we developed the leukosome (LEUKO) coated biomimetic nanoparticles (NPs) for the targeted delivery of Cel to arthritis injury area in RA. LEUKO were synthesized using membrane proteins purified from activated J774 macrophage. LEUKO and Cel-loaded LEUKO (Cel@LEUKO) were characterized using dynamic light scattering and transmission electron microscopy. Our results demonstrated that Cel@LEUKO can inhibit the inflammatory response of lipopolysaccharide (LPS) induced mouse monocyte macrophage leukemia cells (RAW264.7 cells) and human rheumatoid arthritis synovial fibroblasts (MH7A) cells through the inhibition of reactive oxygen species (ROS)-NF-κB pathway. In addition, research has shown that LEUKO effectively targets and transports Cel to the inflammatory site of RA, increased drug concentration in affected areas, reduced systemic toxicity of Cel, and reduced clinical symptoms, inflammatory infiltration, bone erosion, and serum inflammatory factors in collagen-induced arthritis (CIA) rats.
Collapse
Affiliation(s)
- Bo Wang
- Department of Orthopaedics, Suzhou Ruihua Orthopedic Hospital Affiliated Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215000, China; Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, Zhejiang 323000, China.
| | - Jiquan Shen
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, Zhejiang 323000, China
| | - Xinggao Wang
- Department of Orthopaedics, The Sixth Affiliated Hospital of Wenzhou Medical University, The People's Hospital of Lishui, Lishui, Zhejiang 323000, China
| | - Ruixing Hou
- Department of Orthopaedics, Suzhou Ruihua Orthopedic Hospital Affiliated Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
5
|
Yan G, Han Z, Kwon Y, Jousma J, Nukala SB, Prosser BL, Du X, Pinho S, Ong SB, Lee WH, Ong SG. Integrated Stress Response Potentiates Ponatinib-Induced Cardiotoxicity. Circ Res 2024; 134:482-501. [PMID: 38323474 PMCID: PMC10940206 DOI: 10.1161/circresaha.123.323683] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 01/22/2024] [Indexed: 02/08/2024]
Abstract
BACKGROUND Mitochondrial dysfunction is a primary driver of cardiac contractile failure; yet, the cross talk between mitochondrial energetics and signaling regulation remains obscure. Ponatinib, a tyrosine kinase inhibitor used to treat chronic myeloid leukemia, is among the most cardiotoxic tyrosine kinase inhibitors and causes mitochondrial dysfunction. Whether ponatinib-induced mitochondrial dysfunction triggers the integrated stress response (ISR) to induce ponatinib-induced cardiotoxicity remains to be determined. METHODS Using human induced pluripotent stem cells-derived cardiomyocytes and a recently developed mouse model of ponatinib-induced cardiotoxicity, we performed proteomic analysis, molecular and biochemical assays to investigate the relationship between ponatinib-induced mitochondrial stress and ISR and their role in promoting ponatinib-induced cardiotoxicity. RESULTS Proteomic analysis revealed that ponatinib activated the ISR in cardiac cells. We identified GCN2 (general control nonderepressible 2) as the eIF2α (eukaryotic translation initiation factor 2α) kinase responsible for relaying mitochondrial stress signals to trigger the primary ISR effector-ATF4 (activating transcription factor 4), upon ponatinib exposure. Mechanistically, ponatinib treatment exerted inhibitory effects on ATP synthase activity and reduced its expression levels resulting in ATP deficits. Perturbed mitochondrial function resulting in ATP deficits then acts as a trigger of GCN2-mediated ISR activation, effects that were negated by nicotinamide mononucleotide, an NAD+ precursor, supplementation. Genetic inhibition of ATP synthase also activated GCN2. Interestingly, we showed that the decreased abundance of ATP also facilitated direct binding of ponatinib to GCN2, unexpectedly causing its activation most likely because of a conformational change in its structure. Importantly, administering an ISR inhibitor protected human induced pluripotent stem cell-derived cardiomyocytes against ponatinib. Ponatinib-treated mice also exhibited reduced cardiac function, effects that were attenuated upon systemic ISRIB administration. Importantly, ISRIB does not affect the antitumor effects of ponatinib in vitro. CONCLUSIONS Neutralizing ISR hyperactivation could prevent or reverse ponatinib-induced cardiotoxicity. The findings that compromised ATP production potentiates GCN2-mediated ISR activation have broad implications across various cardiac diseases. Our results also highlight an unanticipated role of ponatinib in causing direct activation of a kinase target despite its role as an ATP-competitive kinase inhibitor.
Collapse
Affiliation(s)
- Gege Yan
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Zhenbo Han
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Youjeong Kwon
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Benjamin L Prosser
- Department of Physiology, Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xiaoping Du
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Kowloon Bay, Hong Kong SAR, China
- Kunming Institute of Zoology – The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Neural, Vascular, and Metabolic Biology Thematic Research Program, School of Biomedical Sciences (SBS), Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Won Hee Lee
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, USA
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, USA
| |
Collapse
|
6
|
Gao Y, Ding Y, Tai XR, Zhang C, Wang D. Ponatinib: An update on its drug targets, therapeutic potential and safety. Biochim Biophys Acta Rev Cancer 2023; 1878:188949. [PMID: 37399979 DOI: 10.1016/j.bbcan.2023.188949] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
Leukemia is a malignancy of the hematopoietic system, and as its pathogenesis has become better understood, three generations of tyrosine kinase inhibitors (TKIs) have been developed. Ponatinib is the third-generation breakpoint cluster region (BCR) and Abelson (ABL) TKI, which has been influential in the leukemia therapy for a decade. Moreover, ponatinib is a potent multi-target kinase inhibitor that acts on various kinases, such as KIT, RET, and Src, making it a promising treatment option for triple-negative breast cancer (TNBC), lung cancer, myeloproliferative syndrome, and other diseases. The drug's significant cardiovascular toxicity poses a significant challenge to its clinical use, requiring the development of strategies to minimize its toxicity and side effects. In this article, the pharmacokinetics, targets, therapeutic potential, toxicity and production mechanism of ponatinib will be reviewed. Furthermore, we will discuss methods to reduce the drug's toxicity, providing new avenues for research to improve its safety in clinical use.
Collapse
MESH Headings
- Humans
- Fusion Proteins, bcr-abl/pharmacology
- Fusion Proteins, bcr-abl/therapeutic use
- Drug Resistance, Neoplasm
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/chemically induced
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Antineoplastic Agents/therapeutic use
Collapse
Affiliation(s)
- Yue Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yue Ding
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Xin-Ran Tai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Chen Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Dong Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
7
|
Baudo G, Flinn H, Holcomb M, Tiwari A, Soriano S, Taraballi F, Godin B, Zinger A, Villapol S. Sex-dependent improvement in traumatic brain injury outcomes after liposomal delivery of dexamethasone in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.541045. [PMID: 37292856 PMCID: PMC10245763 DOI: 10.1101/2023.05.16.541045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Traumatic Brain Injury (TBI) can have long-lasting physical, emotional, and cognitive consequences due to the neurodegeneration caused by its robust inflammatory response. Despite advances in rehabilitation care, effective neuroprotective treatments for TBI patients are lacking. Furthermore, current drug delivery methods for TBI treatment are inefficient in targeting inflamed brain areas. To address this issue, we have developed a liposomal nanocarrier (Lipo) encapsulating dexamethasone (Dex), an agonist for the glucocorticoid receptor utilized to alleviate inflammation and swelling in various conditions. In vitro studies show that Lipo-Dex were well tolerated in human and murine neural cells. Lipo-Dex showed significant suppression of inflammatory cytokines, IL-6 and TNF-α, release after induction of neural inflammation with lipopolysaccharide. Further, the Lipo-Dex were administered to young adult male and female C57BL/6 mice immediately after a controlled cortical impact injury. Our findings demonstrate that Lipo-Dex can selectively target the injured brain, thereby reducing lesion volume, cell death, astrogliosis, the release of proinflammatory cytokines, and microglial activation compared to Lipo-treated mice in a sex-dependent manner, showing a major impact only in male mice. This highlights the importance of considering sex as a crucial variable in developing and evaluating new nano-therapies for brain injury. These results suggest that Lipo-Dex administration may effectively treat acute TBI.
Collapse
Affiliation(s)
- Gherardo Baudo
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute Department of Orthopedics and Sports Medicine Houston Methodist Hospital Houston TX, USA
| | - Hannah Flinn
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Morgan Holcomb
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Anjana Tiwari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute Department of Orthopedics and Sports Medicine Houston Methodist Hospital Houston TX, USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Assaf Zinger
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical Engineering, Technion−Israel Institute of Technology, Haifa, Israel
| | - Sonia Villapol
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
8
|
Chen C, Shi Q, Xu J, Ren T, Huang Y, Guo W. Current progress and open challenges for applying tyrosine kinase inhibitors in osteosarcoma. Cell Death Dis 2022; 8:488. [PMID: 36509754 PMCID: PMC9744866 DOI: 10.1038/s41420-022-01252-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/06/2022] [Accepted: 11/10/2022] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OS) is a mesenchymal-origin tumor that constitutes the most common primary malignant bone tumor. The survival rate of the patients has significantly improved since the introduction of neoadjuvant chemotherapy and extensive resection, but it has stagnated in recent 40 years. Tyrosine kinase inhibitors (TKIs) have played a key part in the treatment of malignant tumors. In advanced OS, TKIs including anlotinib, apatinib, sorafenib, etc. have significantly improved the progression-free survival of patients, while the overall survival remains unchanged. The main reason is the rapid and inevitable progress of acquired drug resistance of OS. However, as the application of TKIs in OS and other tumors is still in the exploratory phase, its drug resistance mechanism and corresponding solutions are rarely reported. Hence, in this review, we summarize knowledge of the applications of TKIs, the mechanism of TKIs resistance, and the attempts to overcome TKIs resistance in OS, which are the three potentially novel insights of TKIs in OS. Because most evidence is derived from studies using animal and cell models, we also reviewed clinical trials and related bioinformatics data available in public databases, which partially improved our understanding of TKIs applications.
Collapse
Affiliation(s)
- Chenglong Chen
- grid.414360.40000 0004 0605 7104Department of Orthopedics, Beijing Jishuitan Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Qianyu Shi
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Jiuhui Xu
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Tingting Ren
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Yi Huang
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Wei Guo
- grid.411634.50000 0004 0632 4559Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, People’s Republic of China ,grid.411634.50000 0004 0632 4559Musculoskeletal Tumor Center, Peking University People’s Hospital, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Jampilek J, Kralova K. Insights into Lipid-Based Delivery Nanosystems of Protein-Tyrosine Kinase Inhibitors for Cancer Therapy. Pharmaceutics 2022; 14:2706. [PMID: 36559200 PMCID: PMC9783038 DOI: 10.3390/pharmaceutics14122706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/07/2022] Open
Abstract
According to the WHO, cancer caused almost 10 million deaths worldwide in 2020, i.e., almost one in six deaths. Among the most common are breast, lung, colon and rectal and prostate cancers. Although the diagnosis is more perfect and spectrum of available drugs is large, there is a clear trend of an increase in cancer that ends fatally. A major advance in treatment was the introduction of gentler antineoplastics for targeted therapy-tyrosine kinase inhibitors (TKIs). Although they have undoubtedly revolutionized oncology and hematology, they have significant side effects and limited efficacy. In addition to the design of new TKIs with improved pharmacokinetic and safety profiles, and being more resistant to the development of drug resistance, high expectations are placed on the reformulation of TKIs into various drug delivery lipid-based nanosystems. This review provides an insight into the history of chemotherapy, a brief overview of the development of TKIs for the treatment of cancer and their mechanism of action and summarizes the results of the applications of self-nanoemulsifying drug delivery systems, nanoemulsions, liposomes, solid lipid nanoparticles, lipid-polymer hybrid nanoparticles and nanostructured lipid carriers used as drug delivery systems of TKIs obtained in vitro and in vivo.
Collapse
Affiliation(s)
- Josef Jampilek
- Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska Cesta 9, 845 10 Bratislava, Slovakia
| | - Katarina Kralova
- Institute of Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
10
|
Giordano F, Lenna S, Baudo G, Rampado R, Massaro M, De Rosa E, Ewing A, Kurenbekova L, Agostini M, Yustein JT, Taraballi F. Tyrosine kinase inhibitor-loaded biomimetic nanoparticles as a treatment for osteosarcoma. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00146-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
AbstractSmall-molecule tyrosine kinase inhibitors (TKIs) represent a potentially powerful approach to the treatment of osteosarcoma (OS). However, dose-limiting toxicity, therapeutic efficacy, and targeting specificity are significant barriers to the use of TKIs in the clinic. Notably among TKIs, ponatinib demonstrated potent anti-tumor activity; however, it received an FDA black box warning for potential side effects. We propose ponatinib-loaded biomimetic nanoparticles (NPs) to repurpose ponatinib as an efficient therapeutic option for OS. In this study, we demonstrate enhanced targeting ability and maintain potent ponatinib nano-therapeutic activity, while also reducing toxicity. In in vitro two- and three-dimensional models, we demonstrate that ponatinib-loaded biomimetic NPs maintain the efficacy of the free drug, while in vivo we show that they can improve tumor targeting, slow tumor growth, and reduce evidence of systemic toxicities. Though there is limited Pon encapsulation within NPs, this platform may improve current therapeutic approaches and reduce dosage-related side effects to achieve better clinical outcomes in OS patients.
Graphical Abstract
Collapse
|
11
|
Rampado R, Biccari A, D'Angelo E, Collino F, Cricrì G, Caliceti P, Giordano F, Taraballi F, Pucciarelli S, Agostini M. Optimization of Biomimetic, Leukocyte-Mimicking Nanovesicles for Drug Delivery Against Colorectal Cancer Using a Design of Experiment Approach. Front Bioeng Biotechnol 2022; 10:883034. [PMID: 35757799 PMCID: PMC9214241 DOI: 10.3389/fbioe.2022.883034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/27/2022] [Indexed: 12/22/2022] Open
Abstract
The development of biomimetic nanoparticles (NPs) has revolutionized the concept of nanomedicine by offering a completely new set of biocompatible materials to formulate innovative drug delivery systems capable of imitating the behavior of cells. Specifically, the use of leukocyte-derived membrane proteins to functionalize nanovesicles (leukosomes) can enable their long circulation and target the inflamed endothelium present in many inflammatory pathologies and tumors, making them a promising and versatile drug delivery system. However, these studies did not elucidate the critical experimental parameters involved in leukosomes formulation. In the present study, we approached the preparation of leukosomes using a design of experiment (DoE) method to better understand the influence of experimental parameters on leukosomes features such as size, size distribution, and protein loading. We also validated this formulation technologically and tested its behavior in in vitro colorectal cancer (CRC) models, including CRC patient-derived tumor organoids (PDOs). We demonstrated leukosomes biocompatibility, endothelium adhesion capability, and tumor target in three-dimensional (3D) settings using CRC cell lines. Overall, our study offers a novel conceptual framework for biomimetic NPs using a DoE strategy and consolidates the high therapeutic potential of leukosomes as a viable drug delivery system for anti-inflammatory and antineoplastic applications.
Collapse
Affiliation(s)
- Riccardo Rampado
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nano-Inspired Biomedicine Lab, Institute of Pediatric Research- Città della Speranza, Padua, Italy
| | - Andrea Biccari
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nano-Inspired Biomedicine Lab, Institute of Pediatric Research- Città della Speranza, Padua, Italy
| | - Edoardo D'Angelo
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nano-Inspired Biomedicine Lab, Institute of Pediatric Research- Città della Speranza, Padua, Italy
| | - Federica Collino
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy.,Laboratory of Translational Research in Pediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Giulia Cricrì
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy.,Laboratory of Translational Research in Pediatric Nephro-Urology, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Federica Giordano
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, United States.,Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston, TX, United States.,Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Salvatore Pucciarelli
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy
| | - Marco Agostini
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Padua, Italy.,Nano-Inspired Biomedicine Lab, Institute of Pediatric Research- Città della Speranza, Padua, Italy
| |
Collapse
|
12
|
Rampado R, Caliceti P, Agostini M. Latest Advances in Biomimetic Cell Membrane-Coated and Membrane-Derived Nanovectors for Biomedical Applications. NANOMATERIALS 2022; 12:nano12091543. [PMID: 35564251 PMCID: PMC9104043 DOI: 10.3390/nano12091543] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 01/27/2023]
Abstract
In the last decades, many nanovectors were developed for different diagnostic or therapeutic purposes. However, most nanosystems have been designed using a “bottom-up” approach, in which the basic components of the nanovector become assembled to achieve complex and specific behaviors. Despite the fine control of formulative conditions, the complexity of these systems often results cumbersome and difficult to scale-up. Recently, biomimetic materials emerged as a complementary or alternative design approach through a “top-down strategy”, using cell-derived materials as building blocks to formulate innovative nanovectors. The use of cell membranes as nanoparticle coatings endows nanomaterials with the biological identity and some of the functions of the cells they are derived from. In this review, we discuss some of the latest examples of membrane coated and membrane-derived biomimetic nanomaterials and underline the common general functions offered by the biomaterials used. From these examples, we suggest a systematic classification of these biomimetic materials based on their biological sources and formulation techniques, with their respective advantages and disadvantages, and summarize the current technologies used for membranes isolation and integration on nanovectors. We also discuss some current technical limitations and hint to future direction of the improvement for biomimetics.
Collapse
Affiliation(s)
- Riccardo Rampado
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, 35131 Padua, Italy; (R.R.); (P.C.)
- Nano-Inspired Biomedicine Lab, Insitute of Pediatric Research-Città della Speranza, Corso Stati Uniti 4, 35127 Padua, Italy
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Via Marzolo, 5, 35131 Padua, Italy; (R.R.); (P.C.)
| | - Marco Agostini
- Nano-Inspired Biomedicine Lab, Insitute of Pediatric Research-Città della Speranza, Corso Stati Uniti 4, 35127 Padua, Italy
- General Surgery 3, Department of Surgical, Oncological and Gastroenterological Sciences, University of Padua, Via Nicolò Giustiniani 2, 35128 Padua, Italy
- Correspondence:
| |
Collapse
|
13
|
Chen X, Liu L, Liu P, Chen Y, Lin D, Yan H, Yan Q, Wang Y, Qiu Y, Fang B, Huang H, Qian J, Zhao Y, Du Z, Zhang Q, Li X, Zheng X, Liu Z. Discovery of Potent and Orally Bioavailable Platelet-Derived Growth Factor Receptor (PDGFR) Inhibitors for the Treatment of Osteosarcoma. J Med Chem 2022; 65:5374-5391. [PMID: 35239349 DOI: 10.1021/acs.jmedchem.1c01732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Platelet-derived growth factor receptors (PDGFRs) are now considered promising targets for the treatment of osteosarcoma. Herein, the design, synthesis, and structure-activity relationships (SAR) of novel pyrimidine-2,4-diamine derivatives that selectively inhibit PDGFRα/β kinases have been studied. The screening cascades revealed that 7m was the preferred compound among these derivatives, with IC50 values of 2.4 and 0.9 nM for PDGFRα and PDGFRβ, respectively. Moreover, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) experiment revealed that 7m has a substantial cytotoxic effect against all osteosarcoma cancer cell lines; 7m also displayed robust antitumor effects and low toxicity in a xenograft model. Additionally, 7m showed excellent bioavailability (F = 62.9%), suitable half-life (T1/2 = 2.12 h), satisfactory metabolic stability, and weak CYP isoform inhibitory activity, suggesting that 7m is a potential drug candidate for PDGFR-driven osteosarcoma.
Collapse
Affiliation(s)
- Xiaojing Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Lu Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Peng Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yingying Chen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Dan Lin
- The Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Hao Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Qi Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yi Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yinda Qiu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Bo Fang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Huijing Huang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Jianchang Qian
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yunjie Zhao
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Zhou Du
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Qianwen Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaokun Li
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, People's Republic of China
| |
Collapse
|
14
|
Paradiso F, Lenna S, Gazze SA, Garcia Parra J, Murphy K, Margarit L, Gonzalez D, Francis L, Taraballi F. Mechanomimetic 3D Scaffolds as a Humanized In Vitro Model for Ovarian Cancer. Cells 2022; 11:824. [PMID: 35269446 PMCID: PMC8909508 DOI: 10.3390/cells11050824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
The mechanical homeostasis of tissues can be altered in response to trauma or disease, such as cancer, resulting in altered mechanotransduction pathways that have been shown to impact tumor development, progression, and the efficacy of therapeutic approaches. Specifically, ovarian cancer progression is parallel to an increase in tissue stiffness and fibrosis. With in vivo models proving difficult to study, tying tissue mechanics to altered cellular and molecular properties necessitate advanced, tunable, in vitro 3D models able to mimic normal and tumor mechanic features. First, we characterized normal human ovary and high-grade serous (HGSC) ovarian cancer tissue stiffness to precisely mimic their mechanical features on collagen I-based sponge scaffolds, soft (NS) and stiff (MS), respectively. We utilized three ovarian cancer cell lines (OVCAR-3, Caov-3, and SKOV3) to evaluate changes in viability, morphology, proliferation, and sensitivity to doxorubicin and liposomal doxorubicin treatment in response to a mechanically different microenvironment. High substrate stiffness promoted the proliferation of Caov-3 and SKOV3 cells without changing their morphology, and upregulated mechanosensors YAP/TAZ only in SKOV3 cells. After 7 days in culture, both OVCAR3 and SKOV3 decreased the MS scaffold storage modulus (stiffness), suggesting a link between cell proliferation and the softening of the matrix. Finally, high matrix stiffness resulted in higher OVCAR-3 and SKOV3 cell cytotoxicity in response to doxorubicin. This study demonstrates the promise of biomimetic porous scaffolds for effective inclusion of mechanical parameters in 3D cancer modeling. Furthermore, this work establishes the use of porous scaffolds for studying ovarian cancer cells response to mechanical changes in the microenvironment and as a meaningful platform from which to investigate chemoresistance and drug response.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Stefania Lenna
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - S. Andrea Gazze
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Jezabel Garcia Parra
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Kate Murphy
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK;
| | - Lavinia Margarit
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Deyarina Gonzalez
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Lewis Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
15
|
Giordano F, Lenna S, Rampado R, Brozovich A, Hirase T, Tognon MG, Martini F, Agostini M, Yustein JT, Taraballi F. Nanodelivery Systems Face Challenges and Limitations in Bone Diseases Management. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Federica Giordano
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
| | - Stefania Lenna
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
| | - Riccardo Rampado
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
- First Surgical Clinic Section, Department of Surgical Oncological and Gastroenterological Sciences, University of Padua Padua 35124 Italy
- Nano‐Inspired Biomedicine Laboratory Institute of Pediatric Research—Città della Speranza Padua Italy
| | - Ava Brozovich
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
- Texas A&M College of Medicine 8447 Highway 47 Bryan TX 77807 USA
| | - Takashi Hirase
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
| | - Mauro G. Tognon
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine University of Ferrara Ferrara Italy
| | - Fernanda Martini
- Section of Experimental Medicine, Department of Medical Sciences, School of Medicine University of Ferrara Ferrara Italy
| | - Marco Agostini
- First Surgical Clinic Section, Department of Surgical Oncological and Gastroenterological Sciences, University of Padua Padua 35124 Italy
- Nano‐Inspired Biomedicine Laboratory Institute of Pediatric Research—Città della Speranza Padua Italy
| | - Jason T. Yustein
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center Baylor College of Medicine Houston TX 77030 USA
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration Houston Methodist Academic Institute, Houston Methodist 6670 Bertner Ave Houston TX 77030 USA
- Orthopedics and Sports Medicine Houston Methodist Hospital Houston Methodist, 6565 Fannin Street Houston TX 77030 USA
| |
Collapse
|
16
|
Zinger A, Cvetkovic C, Sushnitha M, Naoi T, Baudo G, Anderson M, Shetty A, Basu N, Covello J, Tasciotti E, Amit M, Xie T, Taraballi F, Krencik R. Humanized Biomimetic Nanovesicles for Neuron Targeting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101437. [PMID: 34382379 PMCID: PMC8498895 DOI: 10.1002/advs.202101437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/09/2021] [Indexed: 05/03/2023]
Abstract
Nanovesicles (NVs) are emerging as innovative, theranostic tools for cargo delivery. Recently, surface engineering of NVs with membrane proteins from specific cell types has been shown to improve the biocompatibility of NVs and enable the integration of functional attributes. However, this type of biomimetic approach has not yet been explored using human neural cells for applications within the nervous system. Here, this paper optimizes and validates the scalable and reproducible production of two types of neuron-targeting NVs, each with a distinct lipid formulation backbone suited to potential therapeutic cargo, by integrating membrane proteins that are unbiasedly sourced from human pluripotent stem-cell-derived neurons. The results establish that both endogenous and genetically engineered cell-derived proteins effectively transfer to NVs without disruption of their physicochemical properties. NVs with neuron-derived membrane proteins exhibit enhanced neuronal association and uptake compared to bare NVs. Viability of 3D neural sphere cultures is not disrupted by treatment, which verifies the utility of organoid-based approaches as NV testing platforms. Finally, these results confirm cellular association and uptake of the biomimetic humanized NVs to neurons within rodent cranial nerves. In summary, the customizable NVs reported here enable next-generation functionalized theranostics aimed to promote neuroregeneration.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal RegenerationHouston Methodist Research InstituteOrthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
- Laboratory for Bioinspired Nano Engineering and Translational Therapeutics, Department of Chemical EngineeringTechnion−Israel Institute of TechnologyHaifa3200003Israel
| | - Caroline Cvetkovic
- Center for NeuroregenerationHouston Methodist Research InstituteDepartment of NeurosurgeryHouston Methodist HospitalHoustonTX77030USA
| | - Manuela Sushnitha
- Center for Musculoskeletal RegenerationHouston Methodist Research InstituteOrthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
- Department of BioengineeringRice UniversityHoustonTX77030USA
| | - Tomoyuki Naoi
- Center for Musculoskeletal RegenerationHouston Methodist Research InstituteOrthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Gherardo Baudo
- Center for Musculoskeletal RegenerationHouston Methodist Research InstituteOrthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Morgan Anderson
- Center for NeuroregenerationHouston Methodist Research InstituteDepartment of NeurosurgeryHouston Methodist HospitalHoustonTX77030USA
| | - Arya Shetty
- Department of BioSciencesRice UniversityHoustonTX77030USA
| | - Nupur Basu
- Center for NeuroregenerationHouston Methodist Research InstituteDepartment of NeurosurgeryHouston Methodist HospitalHoustonTX77030USA
| | - Jennifer Covello
- Department of Head and Neck SurgeryThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | | | - Moran Amit
- Department of Head and Neck SurgeryThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Tongxin Xie
- Department of Head and Neck SurgeryThe University of Texas MD Anderson Cancer CenterHoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Research InstituteOrthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Robert Krencik
- Center for NeuroregenerationHouston Methodist Research InstituteDepartment of NeurosurgeryHouston Methodist HospitalHoustonTX77030USA
| |
Collapse
|
17
|
Zinger A, Soriano S, Baudo G, De Rosa E, Taraballi F, Villapol S. Biomimetic Nanoparticles as a Theranostic Tool for Traumatic Brain Injury. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2100722. [PMID: 34413716 PMCID: PMC8356641 DOI: 10.1002/adfm.202100722] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/08/2021] [Indexed: 05/04/2023]
Abstract
Traumatic brain injury (TBI) triggers both central and peripheral inflammatory responses. Existing pharmacological drugs are unable to effectively and quickly target the brain inflamed regions, setting up a major roadblock towards effective brain trauma treatments. Nanoparticles (NPs) have been used in multiple diseases as drug delivery tools with remarkable success due to their rapid diffusion and specificity in the target organ. Here, leukocyte-based biomimetic NPs are fabricated as a theranostic tool to directly access inflamed regions in a TBI mouse model. This NP systemic delivery is visualized using advanced in vivo imaging techniques, including intravital microscopy and in vivo imaging system. The results demonstrate selective targeting of NPs to the injured brain and increased NPs accumulation among the peripheral organs 24 h after TBI. Interestingly, increased microglial proliferation, decreased macrophage infiltration, and reduced brain lesion following the NPs treatments compared to sham vehicle-treated mice are also found. In summary, the results suggest that NPs represent a promising future theranostic tool for TBI treatment.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sirena Soriano
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
| | - Gherardo Baudo
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Enrica De Rosa
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sonia Villapol
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
- Weill Cornell Medical CollegeNew YorkNY10065USA
| |
Collapse
|
18
|
Russo E, Spallarossa A, Tasso B, Villa C, Brullo C. Nanotechnology of Tyrosine Kinase Inhibitors in Cancer Therapy: A Perspective. Int J Mol Sci 2021; 22:6538. [PMID: 34207175 PMCID: PMC8235113 DOI: 10.3390/ijms22126538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/23/2022] Open
Abstract
Nanotechnology is an important application in modern cancer therapy. In comparison with conventional drug formulations, nanoparticles ensure better penetration into the tumor mass by exploiting the enhanced permeability and retention effect, longer blood circulation times by a reduced renal excretion and a decrease in side effects and drug accumulation in healthy tissues. The most significant classes of nanoparticles (i.e., liposomes, inorganic and organic nanoparticles) are here discussed with a particular focus on their use as delivery systems for small molecule tyrosine kinase inhibitors (TKIs). A number of these new compounds (e.g., Imatinib, Dasatinib, Ponatinib) have been approved as first-line therapy in different cancer types but their clinical use is limited by poor solubility and oral bioavailability. Consequently, new nanoparticle systems are necessary to ameliorate formulations and reduce toxicity. In this review, some of the most important TKIs are reported, focusing on ongoing clinical studies, and the recent drug delivery systems for these molecules are investigated.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| | | | | | | | - Chiara Brullo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| |
Collapse
|
19
|
Zinger A, Sushnitha M, Naoi T, Baudo G, De Rosa E, Chang J, Tasciotti E, Taraballi F. Enhancing Inflammation Targeting Using Tunable Leukocyte-Based Biomimetic Nanoparticles. ACS NANO 2021; 15:6326-6339. [PMID: 33724785 PMCID: PMC8155322 DOI: 10.1021/acsnano.0c05792] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Accepted: 02/22/2021] [Indexed: 05/02/2023]
Abstract
Biomimetic nanoparticles aim to effectively emulate the behavior of either cells or exosomes. Leukocyte-based biomimetic nanoparticles, for instance, incorporate cell membrane proteins to transfer the natural tropism of leukocytes to the final delivery platform. However, tuning the protein integration can affect the in vivo behavior of these nanoparticles and alter their efficacy. Here we show that, while increasing the protein:lipid ratio to a maximum of 1:20 (w/w) maintained the nanoparticle's structural properties, increasing protein content resulted in improved targeting of inflamed endothelium in two different animal models. Our combined use of a microfluidic, bottom-up approach and tuning of a key synthesis parameter enabled the synthesis of reproducible, enhanced biomimetic nanoparticles that have the potential to improve the treatment of inflammatory-based conditions through targeted nanodelivery.
Collapse
Affiliation(s)
- Assaf Zinger
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Manuela Sushnitha
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
- Department
of Bioengineering, Rice University, Houston, Houston, Texas 77030, United States
| | - Tomoyuki Naoi
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Gherardo Baudo
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Enrica De Rosa
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| | - Jenny Chang
- Houston
Methodist Cancer Center, Houston Methodist
Hospital, Houston, Texas 77030, United
States
| | - Ennio Tasciotti
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
- Biotechnology
Program, San Raffaele University and IRCCS
San Raffaele Pisana, 00166 Roma RM, Italy
| | - Francesca Taraballi
- Center
for Musculoskeletal Regeneration, Houston
Methodist Academic Institute, Houston, Texas 77030, United States
- Orthopedics
and Sports Medicine, Houston Methodist Hospital, Houston, Texas 77030, United States
| |
Collapse
|
20
|
Zinger A, Cooke JP, Taraballi F. Biomimetic nano drug delivery carriers for treating cardiovascular diseases. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 33:102360. [PMID: 33476763 DOI: 10.1016/j.nano.2021.102360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 01/02/2023]
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, TX; Orthopedics and Sports Medicine, Houston Methodist Hospital, TX.
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX; Houston Methodist DeBakey Heart and Vascular Center, Houston Methodist Hospital, TX
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, TX; Orthopedics and Sports Medicine, Houston Methodist Hospital, TX.
| |
Collapse
|
21
|
Zinger A, Brozovich A, Pasto A, Sushnitha M, Martinez JO, Evangelopoulos M, Boada C, Tasciotti E, Taraballi F. Bioinspired Extracellular Vesicles: Lessons Learned From Nature for Biomedicine and Bioengineering. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2172. [PMID: 33143238 PMCID: PMC7693812 DOI: 10.3390/nano10112172] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 12/14/2022]
Abstract
Efficient communication is essential in all layers of the biological chain. Cells exchange information using a variety of signaling moieties, such as small molecules, proteins, and nucleic acids. Cells carefully package these messages into lipid complexes, collectively named extracellular vesicles (EVs). In this work, we discuss the nature of these cell carriers, categorize them by their origin, explore their role in the homeostasis of healthy tissues, and examine how they regulate the pathophysiology of several diseases. This review will also address the limitations of using EVs for clinical applications and discuss novel methods to engineer nanoparticles to mimic the structure, function, and features of EVs. Using lessons learned from nature and understanding how cells use EVs to communicate across distant sites, we can develop a better understanding of how to tailor the fundamental features of drug delivery carriers to encapsulate various cargos and target specific sites for biomedicine and bioengineering.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ava Brozovich
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Texas A&M College of Medicine, Bryan, TX 77807, USA
| | - Anna Pasto
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center, 20089 Rozzano, Italy
| | - Manuela Sushnitha
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Bioengineering, Rice University, Houston, TX 77030, USA
| | - Jonathan O. Martinez
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Michael Evangelopoulos
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Christian Boada
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ennio Tasciotti
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
- Biotechnology Program, San Raffaele University, Via di Val Cannuta, 247, 00166 Roma RM, Italy
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.B.); (A.P.); (M.S.); (J.O.M.); (M.E.); (C.B.); (E.T.)
- Department of Orthopedics and Sports Medicine, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|