1
|
Pérez-Lloret M, Reidy E, Lozano-Pérez AA, Marchal JA, Lens PNL, Ryan AE, Erxleben A. Auranofin loaded silk fibroin nanoparticles for colorectal cancer treatment. Drug Deliv Transl Res 2025; 15:1994-2008. [PMID: 39382824 DOI: 10.1007/s13346-024-01719-2] [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] [Accepted: 09/19/2024] [Indexed: 10/10/2024]
Abstract
Colorectal cancer (CRC) is the second most common cause of cancer related deaths worldwide and the prevalence in young people especially is increasing annually. In the search for innovative approaches to treat the disease, drug delivery systems (DDS) are promising owing to their unique properties, which allow improved therapeutic results with lower drug concentrations, overcoming drug resistance and at the same time potentially reducing side effects. Silk fibroin is a biopolymer that can be processed to obtain biocompatible and biodegradable nanoparticles that can be efficiently loaded by surface adsorption with small-molecule therapeutics and allow their transport and sustained release by modulating their pharmacokinetics. Auranofin (AF) has recently been repurposed for its strong anticancer activity and is currently in clinical trials. Its mechanism of action is through the inhibition of thioredoxin reductase enzymes, which play an essential role in several intracellular processes and are overexpressed in some tumours. Taking into account that AF has a low solubility in water, we propose silk fibroin nanoparticles (SFN) as AF carrier in order to improve its bioavailability, increasing cellular absorption and preventing its degradation or avoiding some resistance mechanisms. Here we report the preparation and characterization of a new formulation of AF-loaded silk fibroin nanoparticles (SFN-AF), its functionalization with FITC for the analysis of cellular uptake, as well as its cytotoxic activity against cell lines of human colorectal cancer (HT29 and HCT116) in both 2D and 3D cell cultures. 3D spheroid models provide a 3D environment which mimics the 3D aspects of CRC observed in vivo and represents an effective 3D environment to screen therapeutics for the treatment of CRC. The loaded nanoparticles showed a spherical morphology with a hydrodynamic diameter of ~ 160 nm and good stability in aqueous solution due to their negative surface charges. FESEM-EDX analysis revealed a homogeneous distribution of Au clusters with high electron density on the surface of the nanoparticles. SFN-AF incubated in phosphate buffer at 37 °C released 77% of the loaded AF over 10 days, showing an initial burst and then sustained release. Flow cytometry analysis showed that FITC-SFN-AF was efficiently internalized by both cell lines, which was confirmed by confocal microscopy imaging. SFN enhanced the cytotoxicity of AF in 2D cultures in both CRC lines. Promising results were also obtained in 3D culture paving the way for future application of this strategy as a therapy for CRC.
Collapse
Affiliation(s)
- Marta Pérez-Lloret
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Eileen Reidy
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine Nursing and health Sciences, University of Galway, University Road, Galway, H91TK33, Ireland
- CÚRAM Centre for Medical Devices, University of Galway, Galway, Ireland
- Lambe Institute for Translational Research, School of Medicine, College of Medicine Nursing and health Sciences, University of Galway, Galway, Ireland
| | - Antonio Abel Lozano-Pérez
- Departamento de Biotecnología Genómica y Mejora Vegetal, Instituto Murciano de Investigación y Desarrollo Agrario y Medioambiental, Murcia, 30150, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, Murcia, 30120, Spain
| | - Juan A Marchal
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18016, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs. GRANADA), Granada, 18012, Spain
- Excellence Research Unit Modelling Nature (MNat), University of Granada, Granada, 18016, Spain
- BioFab i3D-Biofabrication and 3D (Bio)Printing Laboratory, University of Granada, Granada, 18100, Spain
| | - Piet N L Lens
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Aideen E Ryan
- Discipline of Pharmacology and Therapeutics, School of Medicine, College of Medicine Nursing and health Sciences, University of Galway, University Road, Galway, H91TK33, Ireland.
- CÚRAM Centre for Medical Devices, University of Galway, Galway, Ireland.
| | - Andrea Erxleben
- School of Biological and Chemical Sciences, University of Galway, University Road, Galway, H91TK33, Ireland.
| |
Collapse
|
2
|
Hoshi R, Gorospe KA, Labouta HI, Azad T, Lee WL, Thu KL. Alternative Strategies for Delivering Immunotherapeutics Targeting the PD-1/PD-L1 Immune Checkpoint in Cancer. Pharmaceutics 2024; 16:1181. [PMID: 39339217 PMCID: PMC11434872 DOI: 10.3390/pharmaceutics16091181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
The programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint constitutes an inhibitory pathway best known for its regulation of cluster of differentiation 8 (CD8)+ T cell-mediated immune responses. Engagement of PD-L1 with PD-1 expressed on CD8+ T cells activates downstream signaling pathways that culminate in T cell exhaustion and/or apoptosis. Physiologically, these immunosuppressive effects exist to prevent autoimmunity, but cancer cells exploit this pathway by overexpressing PD-L1 to facilitate immune escape. Intravenously (IV) administered immune checkpoint inhibitors (ICIs) that block the interaction between PD-1/PD-L1 have achieved great success in reversing T cell exhaustion and promoting tumor regression in various malignancies. However, these ICIs can cause immune-related adverse events (irAEs) due to off-tumor toxicities which limits their therapeutic potential. Therefore, considerable effort has been channeled into exploring alternative delivery strategies that enhance tumor-directed delivery of PD-1/PD-L1 ICIs and reduce irAEs. Here, we briefly describe PD-1/PD-L1-targeted cancer immunotherapy and associated irAEs. We then provide a detailed review of alternative delivery approaches, including locoregional (LDD)-, oncolytic virus (OV)-, nanoparticle (NP)-, and ultrasound and microbubble (USMB)-mediated delivery that are currently under investigation for enhancing tumor-specific delivery to minimize toxic off-tumor effects. We conclude with a commentary on key challenges associated with these delivery methods and potential strategies to mitigate them.
Collapse
Affiliation(s)
- Ryunosuke Hoshi
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Kristyna A. Gorospe
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| | - Hagar I. Labouta
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Leslie Dan Faculty of Pharmacy, University of Toronto, St. George Campus, Toronto, ON M5S 3M2, Canada
- Biomedical Engineering, Faculty of Applied Science and Engineering, University of Toronto, St. George Campus, Toronto, ON M5S 3E2, Canada
| | - Taha Azad
- Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Health Campus, Sherbrooke, QC J1K 2R1, Canada;
- Research Center, Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC J1J 3H5, Canada
| | - Warren L. Lee
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
- Biochemistry, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada
- Medicine and the Interdepartmental Division of Critical Care Medicine, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5B 1T8, Canada
| | - Kelsie L. Thu
- Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, St. George Campus, Toronto, ON M5S 1A8, Canada; (R.H.); (K.A.G.); (W.L.L.)
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada;
| |
Collapse
|
3
|
Shirazi MMA, Saedi TA, Moghaddam ZS, Nemati M, Shiri R, Negahdari B, Goradel NH. Nanotechnology and nano-sized tools: Newer approaches to circumvent oncolytic adenovirus limitations. Pharmacol Ther 2024; 256:108611. [PMID: 38387653 DOI: 10.1016/j.pharmthera.2024.108611] [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: 10/19/2023] [Revised: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Oncolytic adenoviruses (OAds), engineered Ads preferentially infect and lyse tumor cells, have attracted remarkable attention as immunotherapy weapons for the treatment of various malignancies. Despite hopeful successes in preclinical investigations and translation into clinical phases, they face some challenges that thwart their therapeutic effectiveness, including low infectivity of cancer cells, liver sequestration, pre-existing neutralizing antibodies, physical barriers to the spread of Ads, and immunosuppressive TME. Nanotechnology and nano-sized tools provide several advantages to overcome these limitations of OAds. Nano-sized tools could improve the therapeutic efficacy of OAds by enhancing infectivity and cellular uptake, targeting and protecting from pre-existing immune responses, masking and preventing liver tropism, and co-delivery with other therapeutic agents. Herein, we reviewed the constructs of various OAds and their application in clinical trials, as well as the limitations they have faced. Furthermore, we emphasized the potential applications of nanotechnology to solve the constraints of OAds to improve their anti-tumor activities.
Collapse
Affiliation(s)
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| | - Zahra Samadi Moghaddam
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shiri
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran; Arthropod-Borne Diseases Research Centre, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
4
|
Pan X, Ni S, Hu K. Nanomedicines for reversing immunosuppressive microenvironment of hepatocellular carcinoma. Biomaterials 2024; 306:122481. [PMID: 38286109 DOI: 10.1016/j.biomaterials.2024.122481] [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: 09/08/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 01/31/2024]
Abstract
Although immunotherapeutic strategies such as immune checkpoint inhibitors (ICIs) have gained promising advances, their limited efficacy and significant toxicity remain great challenges for hepatocellular carcinoma (HCC) immunotherapy. The tumor immunosuppressive microenvironment (TIME) with insufficient T-cell infiltration and low immunogenicity accounts for most HCC patients' poor response to ICIs. Worse still, the current immunotherapeutics without precise delivery may elicit enormous autoimmune side effects and systemic toxicity in the clinic. With a better understanding of the TIME in HCC, nanomedicines have emerged as an efficient strategy to achieve remodeling of the TIME and superadditive antitumor effects via targeted delivery of immunotherapeutics or multimodal synergistic therapy. Based on the typical characteristics of the TIME in HCC, this review summarizes the recent advancements in nanomedicine-based strategies for TIME-reversing HCC treatment. Additionally, perspectives on the awaiting challenges and opportunities of nanomedicines in modulating the TIME of HCC are presented. Acquisition of knowledge of nanomedicine-mediated TIME reversal will provide researchers with a better opportunity for clinical translation of HCC immunotherapy.
Collapse
Affiliation(s)
- Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Ko J, Song J, Lee Y, Choi N, Kim HN. Understanding organotropism in cancer metastasis using microphysiological systems. LAB ON A CHIP 2024; 24:1542-1556. [PMID: 38192269 DOI: 10.1039/d3lc00889d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Cancer metastasis, the leading cause of cancer-related deaths, remains a complex challenge in medical science. Stephen Paget's "seed and soil theory" introduced the concept of organotropism, suggesting that metastatic success depends on specific organ microenvironments. Understanding organotropism not only offers potential for curbing metastasis but also novel treatment strategies. Microphysiological systems (MPS), especially organ-on-a-chip models, have emerged as transformative tools in this quest. These systems, blending microfluidics, biology, and engineering, grant precise control over cell interactions within organ-specific microenvironments. MPS enable real-time monitoring, morphological analysis, and protein quantification, enhancing our comprehension of cancer dynamics, including tumor migration, vascularization, and pre-metastatic niches. In this review, we explore innovative applications of MPS in investigating cancer metastasis, particularly focusing on organotropism. This interdisciplinary approach converges the field of science, engineering, and medicine, thereby illuminating a path toward groundbreaking discoveries in cancer research.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Jiyoung Song
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Yedam Lee
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
6
|
Sedky NK, Mahdy NK, Abdel-Kader NM, Abdelhady MMM, Maged M, Allam AL, Alfaifi MY, Shamma SN, Hassan HAFM, Fahmy SA. Facile sonochemically-assisted bioengineering of titanium dioxide nanoparticles and deciphering their potential in treating breast and lung cancers: biological, molecular, and computational-based investigations. RSC Adv 2024; 14:8583-8601. [PMID: 38487521 PMCID: PMC10938292 DOI: 10.1039/d3ra08908h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Combining sonochemistry with phytochemistry is a modern trend in the biosynthesis of metallic nanoparticles (NPs), which contributes to the sustainability of chemical processes and minimizes hazardous effects. Herein, titanium dioxide (TiO2) NPs were bioengineered using a novel and facile ultrasound-assisted approach utilizing the greenly extracted essential oil of Ocimum basilicum. FTIR and UV-Vis spectrophotometry were used to confirm the formation of TiO2 NPs. The X-ray diffraction (XRD) analysis showed the crystalline nature of TiO2 NPs. TEM analysis revealed the spherical morphology of the NPs with sizes ranging from 5.55 to 13.89 nm. Energy-dispersive X-ray (EDX) confirmed the purity of the greenly synthesized NPs. TiO2 NPs demonstrated outstanding antitumor activity against breast (MCF-7) and lung (A-549) cancer cells with estimated IC50 values of 1.73 and 4.79 μg mL-1. The TiO2 NPs were cytocompatible to normal cells (MCF-10A) with a selectivity index (SI) of 8.77 for breast and 3.17 for lung cancer. Biological assays revealed a promising potential for TiO2 NPs to induce apoptosis and arrest cells at the sub-G1 phase of the cell cycle phase in both cancer cell lines. Molecular investigations showed the ability of TiO2 NPs to increase apoptotic genes' expression (Bak and Bax) and their profound ability to elevate the expression of apoptotic proteins (caspases 3 and 7). Molecular docking demonstrated strong binding interactions for TiO2 NPs with caspase 3 and EGFR-TK targets. In conclusion, the greenly synthesized TiO2 NPs exhibited potent antitumor activity and mitochondrion-based cell death against breast and lung cancer cell lines while maintaining cytocompatibility against normal cells.
Collapse
Affiliation(s)
- Nada K Sedky
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
| | - Noha Khalil Mahdy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
| | - Nour M Abdel-Kader
- Department of Biochemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Administrative Capital Cairo 11835 Egypt
- Department of Biochemistry, Faculty of Science, Ain Shams University Cairo 11566 Egypt
| | - Manal M M Abdelhady
- Clinical Pharmacy Department, Faculty of Pharmacy, Badr University Cairo 11829 Egypt
| | - Mohamad Maged
- Faculty of Biotechnology, Nile University Giza Egypt
| | - Aya L Allam
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Egypt
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
| | - Samir N Shamma
- Institute of Global Health and Human Ecology, School of Sciences & Engineering, The American University in Cairo AUC Avenue, P.O. Box 74 New Cairo 11835 Egypt
| | - Hatem A F M Hassan
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Egypt
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University 11562 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Chemistry, School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation R5 New Garden City, New Capital Cairo 11835 Egypt +20-1222613344
| |
Collapse
|
7
|
Tang B, Ma W, Lin Y. Emerging applications of anti-angiogenic nanomaterials in oncotherapy. J Control Release 2023; 364:61-78. [PMID: 37871753 DOI: 10.1016/j.jconrel.2023.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 10/25/2023]
Abstract
Angiogenesis is the process of generating new blood vessels from pre-existing vasculature. Under normal conditions, this process is delicately controlled by pro-angiogenic and anti-angiogenic factors. Tumor cells can produce plentiful pro-angiogenic molecules promoting pathological angiogenesis for uncontrollable growth. Therefore, anti-angiogenic therapy, which aims to inhibit tumor angiogenesis, has become an attractive approach for oncotherapy. However, classic anti-angiogenic agents have several limitations in clinical use, such as lack of specific targeting, low bioavailability, and poor therapeutic outcomes. Hence, alternative angiogenic inhibitors are highly desired. With the emergence of nanotechnology, various nanomaterials have been designed for anti-angiogenesis purposes, offering promising features like excellent targeting capabilities, reduced side effects, and enhanced therapeutic efficacy. In this review, we describe tumor vascular features, discuss current dilemma of traditional anti-angiogenic medicines in oncotherapy, and underline the potential of nanomaterials in tumor anti-angiogenic therapy. Moreover, we discuss the current challenges of anti-angiogenic cancer treatment. We expect that this summary of anti-angiogenic nanomaterials in oncotherapy will offer valuable insights, facilitating their extensive applications in the future.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China; Sichuan Provincial Engineering Research Center of Oral Biomaterials, Chengdu, Sichuan 610041, China; Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
8
|
Becker S, Momoh J, Biancacci I, Möckel D, Wang Q, May JN, Su H, Candels LS, Berres ML, Kiessling F, Hatting M, Lammers T, Trautwein C. Intermittent Fasting Primes the Tumor Microenvironment and Improves Nanomedicine Delivery in Hepatocellular Carcinoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2208042. [PMID: 37376850 DOI: 10.1002/smll.202208042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/18/2023] [Indexed: 06/29/2023]
Abstract
Fasting has many health benefits, including reduced chemotherapy toxicity and improved efficacy. It is unclear how fasting affects the tumor microenvironment (TME) and tumor-targeted drug delivery. Here the effects of intermittent (IF) and short-term (STF) fasting are investigated on tumor growth, TME composition, and liposome delivery in allogeneic hepatocellular carcinoma (HCC) mouse models. To this end, mice are inoculated either subcutaneously or intrahepatically with Hep-55.1C cells and subjected to IF for 24 d or to STF for 1 d. IF but not STF significantly slows down tumor growth. IF increases tumor vascularization and decreases collagen density, resulting in improved liposome delivery. In vitro, fasting furthermore promotes the tumor cell uptake of liposomes. These results demonstrate that IF shapes the TME in HCC towards enhanced drug delivery. Finally, when combining IF with liposomal doxorubicin treatment, the antitumor efficacy of nanochemotherapy is found to be increased, while systemic side effects are reduced. Altogether, these findings exemplify that the beneficial effects of fasting on anticancer therapy outcomes go beyond modulating metabolism at the molecular level.
Collapse
Affiliation(s)
- Svea Becker
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jeffrey Momoh
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Ilaria Biancacci
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Diana Möckel
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Qingbi Wang
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Jan-Niklas May
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Huan Su
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Lena Susanna Candels
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Marie-Luise Berres
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Maximilian Hatting
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), University Hospital RWTH Aachen, 52074, Aachen, Germany
| | - Christian Trautwein
- Clinic for Gastroenterology, Metabolic Disorders, and Internal Intensive Medicine (Med III), University Hospital RWTH Aachen, 52074, Aachen, Germany
| |
Collapse
|
9
|
Lahooti B, Akwii RG, Zahra FT, Sajib MS, Lamprou M, Alobaida A, Lionakis MS, Mattheolabakis G, Mikelis CM. Targeting endothelial permeability in the EPR effect. J Control Release 2023; 361:212-235. [PMID: 37517543 DOI: 10.1016/j.jconrel.2023.07.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 08/01/2023]
Abstract
The characteristics of the primary tumor blood vessels and the tumor microenvironment drive the enhanced permeability and retention (EPR) effect, which confers an advantage towards enhanced delivery of anti-cancer nanomedicine and has shown beneficial effects in preclinical models. Increased vascular permeability is a landmark feature of the tumor vessels and an important driver of the EPR. The main focus of this review is the endothelial regulation of vascular permeability. We discuss current challenges of targeting vascular permeability towards clinical translation and summarize the structural components and mechanisms of endothelial permeability, the principal mediators and signaling players, the targeted approaches that have been used and their outcomes to date. We also critically discuss the effects of the tumor-infiltrating immune cells, their interplay with the tumor vessels and the impact of immune responses on nanomedicine delivery, the impact of anti-angiogenic and tumor-stroma targeting approaches, and desirable nanoparticle design approaches for greater translational benefit.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Fatema Tuz Zahra
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Md Sanaullah Sajib
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Margarita Lamprou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
| | - Ahmed Alobaida
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il 81442, Saudi Arabia
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece.
| |
Collapse
|
10
|
Izci M, Maksoudian C, Gonçalves F, Pérez Gilabert I, Rios Luci C, Bolea-Fernandez E, Vanhaecke F, Manshian BB, Soenen SJ. The Efficacy of Nanoparticle Delivery to Hypoxic Solid Tumors by ciRGD Co-Administration Depends on Neuropilin-1 and Neutrophil Levels. Adv Healthc Mater 2023; 12:e2300594. [PMID: 37247322 DOI: 10.1002/adhm.202300594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/02/2023] [Indexed: 05/31/2023]
Abstract
The ability to improve nanoparticle delivery to solid tumors is an actively studied domain, where various mechanisms are looked into. In previous work, the authors have looked into nanoparticle size, tumor vessel normalization, and disintegration, and here it is aimed to continue this work by performing an in-depth mechanistic study on the use of ciRGD peptide co-administration. Using a multiparametric approach, it is observed that ciRGD can improve nanoparticle delivery to the tumor itself, but also to tumor cells specifically better than vessel normalization strategies. The effect depends on the level of tumor perfusion, hypoxia, neutrophil levels, and vessel permeability. This work shows that upon characterizing tumors for these parameters, conditions can be selected that can optimally benefit from ciRGD co-administration as a means to improve NP delivery to solid tumors.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Christy Maksoudian
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Filipa Gonçalves
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Irati Pérez Gilabert
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Carla Rios Luci
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
| | - Eduardo Bolea-Fernandez
- Atomic & Mass Spectrometry - A&MS research group, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, Ghent, 9000, Belgium
| | - Frank Vanhaecke
- Atomic & Mass Spectrometry - A&MS research group, Department of Chemistry, Ghent University, Campus Sterre, Krijgslaan 281-S12, Ghent, 9000, Belgium
| | - Bella B Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
- Leuven Cancer Research Institute, Faculty of Medical Sciences, KU Leuven, Herestraat 49, Leuven, B3000, Belgium
| | - Stefaan J Soenen
- NanoHealth and Optical Imaging Group, Department of Imaging and Pathology, KU Leuven Herestraat 49, Leuven, B3000, Belgium
- Leuven Cancer Research Institute, Faculty of Medical Sciences, KU Leuven, Herestraat 49, Leuven, B3000, Belgium
| |
Collapse
|
11
|
Sun XX, Nosrati Z, Ko J, Lee CM, Bennewith KL, Bally MB. Induced Vascular Normalization-Can One Force Tumors to Surrender to a Better Microenvironment? Pharmaceutics 2023; 15:2022. [PMID: 37631236 PMCID: PMC10458586 DOI: 10.3390/pharmaceutics15082022] [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: 05/03/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy has changed the way many cancers are being treated. Researchers in the field of immunotherapy and tumor immunology are investigating similar questions: How can the positive benefits achieved with immunotherapies be enhanced? Can this be achieved through combinations with other agents and if so, which ones? In our view, there is an urgent need to improve immunotherapy to make further gains in the overall survival for those patients that should benefit from immunotherapy. While numerous different approaches are being considered, our team believes that drug delivery methods along with appropriately selected small-molecule drugs and drug candidates could help reach the goal of doubling the overall survival rate that is seen in some patients that are given immunotherapeutics. This review article is prepared to address how immunotherapies should be combined with a second treatment using an approach that could realize therapeutic gains 10 years from now. For context, an overview of immunotherapy and cancer angiogenesis is provided. The major targets in angiogenesis that have modulatory effects on the tumor microenvironment and immune cells are highlighted. A combination approach that, for us, has the greatest potential for success involves treatments that will normalize the tumor's blood vessel structure and alter the immune microenvironment to support the action of immunotherapeutics. So, this is reviewed as well. Our focus is to provide an insight into some strategies that will engender vascular normalization that may be better than previously described approaches. The potential for drug delivery systems to promote tumor blood vessel normalization is considered.
Collapse
Affiliation(s)
- Xu Xin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Zeynab Nosrati
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Janell Ko
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
| | - Che-Min Lee
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kevin L. Bennewith
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
12
|
Li Y, Si R, Wang J, Hai P, Zheng Y, Zhang Q, Pan X, Zhang J. Discovery of novel antibody-drug conjugates bearing tissue protease specific linker with both anti-angiogenic and strong cytotoxic effects. Bioorg Chem 2023; 137:106575. [PMID: 37148706 DOI: 10.1016/j.bioorg.2023.106575] [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: 12/18/2022] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
Bevacizumab is an FDA-approved class of monoclonal antibodies used to inhibit angiogenesis and promote normalization of blood vessels. It is usually combined with chemotherapeutic agents to treat a variety of solid tumors. However, the whole-body toxicities and toxicity associated with chemotherapy greatly limit the clinical use of this combination therapy. Antibody-drug conjugates (ADCs) couple monoclonal antibodies to cytotoxic molecules via a linker, utilizing the high specificity of monoclonal antibodies to tumor surface antigens to act as a "biological missile" to deliver chemotherapeutic drugs to the tumor site. Herein, we designed a bevacizumab-based ADC, Bevacizumab Vedotin, conjugating bevacizumab to the microtubulin inhibitor MMAE via a tissue protease-specific linker. Biological studies showed strong stability and good tumor cell targeting of our constructed ADCs; rapid drug release was achieved in the presence of exogenous histone protease B. In addition, Bevacizumab Vedotin exhibited good anti-proliferative, apoptosis-promoting and cell cycle-stalling effects on glioma (U87), hepatocellular carcinoma (HepG2), and breast cancer (MCF-7) cell lines. Further in vitro assays demonstrated the enhanced anti-migration activity against MCF-7, potent anti-angiogenic effects, and blockade of the VEGF/VEGFR pathway of Bevacizumab Vedotin.
Collapse
Affiliation(s)
- Yanchen Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ru Si
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Hai
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining 810016, China
| | - Yongbiao Zheng
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining 810016, China
| | - Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
13
|
Lahooti B, Akwii RG, Patel D, ShahbaziNia S, Lamprou M, Madadi M, Abbruscato TJ, Astrinidis A, Bickel U, Al-Ahmad A, German NA, Mattheolabakis G, Mikelis CM. Endothelial-Specific Targeting of RhoA Signaling via CD31 Antibody-Conjugated Nanoparticles. J Pharmacol Exp Ther 2023; 385:35-49. [PMID: 36746610 PMCID: PMC10029826 DOI: 10.1124/jpet.122.001384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/04/2023] [Accepted: 01/17/2023] [Indexed: 02/08/2023] Open
Abstract
Existing vascular endothelial growth factor-oriented antiangiogenic approaches are known for their high potency. However, significant side effects associated with their use drive the need for novel antiangiogenic strategies. The small GTPase RhoA is an established regulator of actin cytoskeletal dynamics. Previous studies have highlighted the impact of endothelial RhoA pathway on angiogenesis. Rho-associate kinase (ROCK), a direct RhoA effector, is potently inhibited by Fasudil, a clinically relevant ROCK inhibitor. Here, we aimed to target the RhoA signaling in endothelial cells by generating Fasudil-encapsulated CD31-targeting liposomes as a potential antiangiogenic therapy. The liposomes presented desirable characteristics, preferential binding to CD31-expressing HEK293T cells and to endothelial cells, inhibited stress fiber formation and cytoskeletal-related morphometric parameters, and inhibited in vitro angiogenic functions. Overall, this work shows that the nanodelivery-mediated endothelial targeting of RhoA signaling can offer a promising strategy for angiogenesis inhibition in vascular-related diseases. SIGNIFICANCE STATEMENT: Systemic administration of antiangiogenic therapeutics induces side effects to non-targeted tissues. This study, among others, has shown the impact of the RhoA signaling in the endothelial cells and their angiogenic functions. Here, to minimize potential toxicity, this study generated CD31-targeting liposomes with encapsulated Fasudil, a clinically relevant Rho kinase inhibitor, and successfully targeted endothelial cells. In this proof-of-principle study, the efficient Fasudil delivery, its impact on the endothelial signaling, morphometric alterations, and angiogenic functions verify the benefits of site-targeted antiangiogenic therapy.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Racheal G Akwii
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Siavash ShahbaziNia
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Margarita Lamprou
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Mahboubeh Madadi
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Aristotelis Astrinidis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Ulrich Bickel
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Abraham Al-Ahmad
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Nadezhda A German
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - George Mattheolabakis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| | - Constantinos M Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas (B.L., R.G.A., D.P., S.S., T.J.A., U.B., A.A.-A., N.A.G., C.M.M.); Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras, Greece(M.L., C.M.M.); Department of Marketing and Business Analytics, Lucas College and Graduate School of Business, San Jose State University, San Jose, California (M.M.); Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital, Memphis, Tennessee (A.A.); and School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana (G.M.)
| |
Collapse
|
14
|
Bahreyni A, Mohamud Y, Luo H. Recent advancements in immunotherapy of melanoma using nanotechnology-based strategies. Biomed Pharmacother 2023; 159:114243. [PMID: 36641926 DOI: 10.1016/j.biopha.2023.114243] [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: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Melanoma is a malignant tumor that accounts for the deadliest form of skin cancers. Despite the significant efforts made recently for development of immunotherapeutic strategies including using immune checkpoint inhibitors and cancer vaccines, the clinical outcomes are unsatisfying. Different factors affect efficient cancer immunotherapy such as side-effects, immunosuppressive tumor microenvironment, and tumor heterogeneity. In the past decades, various nanotechnology-based approaches have been developed to enhance the efficacy of cancer immunotherapy, in addition to diminishing the toxicity associated with it. Several studies have shown that proper application of nanomaterials can revolutionize the outcome of immunotherapy in diverse melanoma models. This review summarizes the recent advancement in the integration of nanotechnology and cancer immunotherapy in melanoma treatment. The importance of nanomaterials and their therapeutic advantages for patients with melanoma are also discussed.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada; Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada.
| |
Collapse
|
15
|
Rakhilin N, Yang B, Spilker ME, Manzuk LK, Montgomery MK, Shin E, Prashad N, Hwang J, Song Y, Loganzo F, Giddabasappa A, Ram S. Volumetric imaging of optically cleared and fluorescently labeled animal tissue (VIOLA) for quantifying the 3D biodistribution of nanoparticles at cellular resolution in tumor tissue. J Control Release 2023; 354:244-259. [PMID: 36596340 DOI: 10.1016/j.jconrel.2022.12.060] [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: 09/23/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Nanoparticle (NP) technology holds significant promise to mediate targeted drug delivery to specific organs in the body. Understanding the 3D biodistribution of NPs in heterogeneous environments such as the tumor tissue can provide crucial information on efficacy, safety and potential clinical outcomes. Here we present a novel end-to-end workflow, VIOLA, which makes use of tissue clearing methodology in conjunction with high resolution imaging and advanced 3D image processing to quantify the spatiotemporal 3D biodistribution of fluorescently labeled ACCURIN® NPs. Specifically, we investigate the spatiotemporal biodistribution of NPs in three different murine tumor models (CT26, EMT6, and KPC-GEM) of increasing complexity and translational relevance. We have developed new endpoints to characterize NP biodistribution at multiple length scales. Our observations reveal that the macroscale NP biodistribution is spatially heterogeneous and exhibits a gradient with relatively high accumulation at the tumor periphery that progressively decreases towards the tumor core in all the tumor models. Microscale analysis revealed that NP extravasation from blood vessels increases in a time dependent manner and plateaus at 72 h post injection. Volumetric analysis and pharmacokinetic modeling of NP biodistribution in the vicinity of the blood vessels revealed that the local NP density exhibits a distance dependent spatiotemporal biodistribution which provide insights into the dynamics of NP extravasation in the tumor tissue. Our data represents a comprehensive analysis of NP biodistribution at multiple length scales in different tumor models providing unique insights into their spatiotemporal dynamics. Specifically, our results show that NPs exhibit a dynamic equilibrium with macroscale heterogeneity combined with microscale homogeneity.
Collapse
Affiliation(s)
| | - Bing Yang
- Comparative Medicine, Pfizer Inc., United States
| | - Mary E Spilker
- Medicine Design - Translational Modeling and Simulation, Pfizer Inc., United States
| | | | | | - Eyoung Shin
- Oncology Research Unit, Pfizer Inc., United States
| | | | | | - Youngho Song
- Oncology Research Unit, Pfizer Inc., United States
| | | | | | - Sripad Ram
- Drug Safety R&D, Pfizer Inc., United States.
| |
Collapse
|
16
|
Luo Y, Wang J, Xu L, Du Q, Fang N, Wu H, Liu F, Hu L, Xu J, Hou J, Zhong Y, Liu Y, Wang Z, Ran H, Guo D. A theranostic metallodrug modulates immunovascular crosstalk to combat immunosuppressive liver cancer. Acta Biomater 2022; 154:478-496. [PMID: 36280029 DOI: 10.1016/j.actbio.2022.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/23/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Hepatocellular carcinoma (HCC) is a highly malignant, fatal disease with a complex tumor microenvironment (TME) characterized by severe immunosuppression and malformed vascular structures, thus most advanced HCC patients do not respond well to current mainstream pharmacotherapy and T-cell-related immunotherapy. Therefore, an efficient immunovascular crosstalk modulation strategy may help combat HCC by reversing immunosuppression and vessel normalization, especially by reprogramming tumor associated macrophages (TAMs). In this study, tyrosine kinase inhibitor lenvatinib (Len) was loaded into mesoporous Fe3O4 (mFe) nanoparticles (NPs), and bovine serum albumin (BSA) was attached to the NP surface to produce a metallodrug (BSA-mFe@Len NPs). In acidic TME, BSA allowed pH-responsive Len release and mFe exposure. Len directly triggered HCC apoptosis and changed the abnormal TME via vessel normalization, cytotoxic T-lymphocyte recruitment, and regulatory T-cell elimination at tailored dosages. After TAM phagocytosis, mFe NPs reprogrammed TAMs into M1 phenotypes to synergistically amplify antitumor immunity. The metallodrug achieved significant tumor growth inhibition, induced tumor vessel normalization effects, and acquired instant antitumor immunity as well as long-term immune memory in vivo. Furthermore, it displayed good T2 weighted magnetic resonance imaging performance, indicating potential theranostic applications. Collectively, this research provides new insights for unleashing the multifaceted potential of current pharmaceuticals in synergy with metallic nanomedicine for treating intractable liver cancer. STATEMENT OF SIGNIFICANCE: Current pharmacotherapy and immunotherapy have limited success in treating advanced hepatocellular carcinoma (HCC) due to its complex tumor microenvironment (TME). Hence, this work first put forward a theranostic metallodrug by loading lenvatinib (Len) into mesoporous Fe3O4 (mFe) nanoparticles (NPs) and coating a pH-degradable bovine serum albumin corona onto the surface. The metallodrug was able to modulate immunovascular TME for combating HCC via metalloimmunotherapy induced by the mFe NPs and Len's multiple functions (direct triggering of tumor apoptosis, vessel normalization, cytotoxic T-lymphocyte recruitment, and regulatory T-cell elimination). In vivo experiments showed that the metallodrug could significantly inhibit HCC growth and evoke long-term antitumor immune memory, paving a new avenue for treating advanced HCC patients.
Collapse
Affiliation(s)
- Ying Luo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Junrui Wang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Lian Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Qianying Du
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Ni Fang
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Hongyun Wu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Fan Liu
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Liu Hu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Jie Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Jingxin Hou
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yixin Zhong
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China; Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China
| | - Dajing Guo
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, PR China.
| |
Collapse
|
17
|
Xiao M, Shi Y, Jiang S, Cao M, Chen W, Xu Y, Xu Z, Wang K. Recent advances of nanomaterial-based anti-angiogenic therapy in tumor vascular normalization and immunotherapy. Front Oncol 2022; 12:1039378. [PMID: 36523993 PMCID: PMC9745116 DOI: 10.3389/fonc.2022.1039378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/14/2022] [Indexed: 01/04/2025] Open
Abstract
Anti-angiogenesis therapy and immunotherapy are the first-line therapeutic strategies for various tumor treatments in the clinic, bringing significant advantages for tumor patients. Recent studies have shown that anti-angiogenic therapy can potentiate immunotherapy, with many clinical trials conducted based on the combination of anti-angiogenic agents and immune checkpoint inhibitors (ICIs). However, currently available clinical dosing strategies and tools are limited, emphasizing the need for more improvements. Although significant progress has been achieved, several big questions remained, such as how to achieve cell-specific targeting in the tumor microenvironment? How to improve drug delivery efficiency in tumors? Can nanotechnology be used to potentiate existing clinical drugs and achieve synergistic sensitization effects? Over the recent few years, nanomedicines have shown unique advantages in antitumor research, including cell-specific targeting, improved delivery potentiation, and photothermal effects. Given that the applications of nanomaterials in tumor immunotherapy have been widely reported, this review provides a comprehensive overview of research advances on nanomaterials in anti-angiogenesis therapy, mainly focusing on the immunosuppressive effects of abnormal tumor vessels in the tumor immune microenvironment, the targets and strategies of anti-angiogenesis nanomedicines, and the potential synergistic effects and molecular mechanisms of anti-angiogenic nanomedicines in combination with immunotherapy, ultimately providing new perspectives on the nanomedicine-based synergy between anti-angiogenic and immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
| |
Collapse
|
18
|
Enhanced Permeability and Retention Effect as a Ubiquitous and Epoch-Making Phenomenon for the Selective Drug Targeting of Solid Tumors. J Pers Med 2022; 12:jpm12121964. [PMID: 36556185 PMCID: PMC9784116 DOI: 10.3390/jpm12121964] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
In 1979, development of the first polymer drug SMANCS [styrene-co-maleic acid (SMA) copolymer conjugated to neocarzinostatin (NCS)] by Maeda and colleagues was a breakthrough in the cancer field. When SMANCS was administered to mice, drug accumulation in tumors was markedly increased compared with accumulation of the parental drug NCS. This momentous result led to discovery of the enhanced permeability and retention effect (EPR effect) in 1986. Later, the EPR effect became known worldwide, especially in nanomedicine, and is still believed to be a universal mechanism for tumor-selective accumulation of nanomedicines. Some research groups recently characterized the EPR effect as a controversial concept and stated that it has not been fully demonstrated in clinical settings, but this erroneous belief is due to non-standard drug design and use of inappropriate tumor models in investigations. Many research groups recently provided solid evidence of the EPR effect in human cancers (e.g., renal and breast), with significant diversity and heterogeneity in various patients. In this review, we focus on the dynamics of the EPR effect and restoring tumor blood flow by using EPR effect enhancers. We also discuss new applications of EPR-based nanomedicine in boron neutron capture therapy and photodynamic therapy for solid tumors.
Collapse
|
19
|
Ileiwat ZE, Tabish TA, Zinovkin DA, Yuzugulen J, Arghiani N, Pranjol MZI. The mechanistic immunosuppressive role of the tumour vasculature and potential nanoparticle-mediated therapeutic strategies. Front Immunol 2022; 13:976677. [PMID: 36045675 PMCID: PMC9423123 DOI: 10.3389/fimmu.2022.976677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022] Open
Abstract
The tumour vasculature is well-established to display irregular structure and hierarchy that is conducive to promoting tumour growth and metastasis while maintaining immunosuppression. As tumours grow, their metabolic rate increases while their distance from blood vessels furthers, generating a hypoxic and acidic tumour microenvironment. Consequently, cancer cells upregulate the expression of pro-angiogenic factors which propagate aberrant blood vessel formation. This generates atypical vascular features that reduce chemotherapy, radiotherapy, and immunotherapy efficacy. Therefore, the development of therapies aiming to restore the vasculature to a functional state remains a necessary research target. Many anti-angiogenic therapies aim to target this such as bevacizumab or sunitinib but have shown variable efficacy in solid tumours due to intrinsic or acquired resistance. Therefore, novel therapeutic strategies such as combination therapies and nanotechnology-mediated therapies may provide alternatives to overcoming the barriers generated by the tumour vasculature. This review summarises the mechanisms that induce abnormal tumour angiogenesis and how the vasculature’s features elicit immunosuppression. Furthermore, the review explores examples of treatment regiments that target the tumour vasculature.
Collapse
Affiliation(s)
- Zakaria Elias Ileiwat
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Tanveer A. Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nahid Arghiani
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- *Correspondence: Nahid Arghiani, ; Md Zahidul I. Pranjol,
| | - Md Zahidul I. Pranjol
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- *Correspondence: Nahid Arghiani, ; Md Zahidul I. Pranjol,
| |
Collapse
|
20
|
He J, Liu Z, Zhu X, Xia H, Gao H, Lu J. Ultrasonic Microbubble Cavitation Enhanced Tissue Permeability and Drug Diffusion in Solid Tumor Therapy. Pharmaceutics 2022; 14:1642. [PMID: 36015267 PMCID: PMC9414228 DOI: 10.3390/pharmaceutics14081642] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/31/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy has an essential role not only in advanced solid tumor therapy intervention but also in society's health at large. Chemoresistance, however, seriously restricts the efficiency and sensitivity of chemotherapeutic agents, representing a significant threat to patients' quality of life and life expectancy. How to reverse chemoresistance, improve efficacy sensitization response, and reduce adverse side effects need to be tackled urgently. Recently, studies on the effect of ultrasonic microbubble cavitation on enhanced tissue permeability and retention (EPR) have attracted the attention of researchers. Compared with the traditional targeted drug delivery regimen, the microbubble cavitation effect, which can be used to enhance the EPR effect, has the advantages of less trauma, low cost, and good sensitization effect, and has significant application prospects. This article reviews the research progress of ultrasound-mediated microbubble cavitation in the treatment of solid tumors and discusses its mechanism of action to provide new ideas for better treatment strategies.
Collapse
Affiliation(s)
- Jide He
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Zenan Liu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Xuehua Zhu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
21
|
Awasthi N, Schwarz MA, Zhang C, Klinz SG, Meyer-Losic F, Beaufils B, Thiagalingam A, Schwarz RE. Augmenting Experimental Gastric Cancer Activity of Irinotecan through Liposomal Formulation and Antiangiogenic Combination Therapy. Mol Cancer Ther 2022; 21:1149-1159. [PMID: 35500018 PMCID: PMC9377761 DOI: 10.1158/1535-7163.mct-21-0860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/22/2022] [Accepted: 04/28/2022] [Indexed: 01/07/2023]
Abstract
Gastric adenocarcinoma (GAC) is the third most common cause of cancer-related deaths worldwide. Combination chemotherapy remains the standard treatment for advanced GAC. Liposomal irinotecan (nal-IRI) has improved pharmacokinetics (PK) and drug biodistribution compared with irinotecan (IRI, CPT-11). Angiogenesis plays a crucial role in the progression and metastasis of GAC. We evaluated the antitumor efficacy of nal-IRI in combination with novel antiangiogenic agents in GAC mouse models. Animal survival studies were performed in peritoneal dissemination xenografts. Tumor growth and PK studies were performed in subcutaneous xenografts. Compared with controls, extension in animal survival by nal-IRI and IRI was >156% and >94%, respectively. The addition of nintedanib or DC101 extended nal-IRI response by 13% and 15%, and IRI response by 37% and 31% (MKN-45 xenografts); nal-IRI response by 11% and 3%, and IRI response by 16% and 40% (KATO-III xenografts). Retardation of tumor growth was greater with nal-IRI (92%) than IRI (71%). Nintedanib and DC101 addition tend to augment nal-IRI or IRI response in this model. The addition of antiangiogenic agents enhanced tumor cell proliferation inhibition effects of nal-IRI or IRI. The tumor vasculature was decreased by nintedanib (65%) and DC101 (58%), while nal-IRI and IRI alone showed no effect. PK characterization in GAC xenografts demonstrated that compared with IRI, nal-IRI treatment groups had higher retention, circulation time, and tumor levels of CPT-11 and its active metabolite SN-38. These findings indicate that nal-IRI, alone and in combination with antiangiogenic agents, has the potential for improving clinical GAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | - Margaret A. Schwarz
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
- Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| | - Changhua Zhang
- Department of Gastrointestinal Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Guangming, Shenzhen, China
| | | | | | | | | | - Roderich E. Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, Indiana
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| |
Collapse
|
22
|
Pancewicz J, Niklińska WE, Chlanda A. Flake Graphene-Based Nanomaterial Approach for Triggering a Ferroptosis as an Attractive Theranostic Outlook for Tackling Non-Small Lung Cancer: A Mini Review. MATERIALS (BASEL, SWITZERLAND) 2022; 15:3456. [PMID: 35629488 PMCID: PMC9143918 DOI: 10.3390/ma15103456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Lung cancer is a highly aggressive neoplasm that is now a leading cause of cancer death worldwide. One of the major approaches for killing cancer cells is related with activation of apoptotic cell death with anti-cancer drugs. However, the efficiency of apoptosis induction in tumors is limited. Consequently, the development of other forms of non-apoptotic cell death is up to date challenge for scientists worldwide. This situation motivated us to define the aim of this mini-review: gathering knowledge regarding ferroptosis-newly defined programmed cell death process characterized by the excessive accumulation of iron-and combining it with yet another interesting nanomaterial-based graphene approach. In this manuscript, we presented brief information about non-small lung cancer and ferroptosis, followed by a section depicting the key-features of graphene-based nanomaterials influencing their biologically relevant properties.
Collapse
Affiliation(s)
- Joanna Pancewicz
- Department of Histology and Embryology, Medical University in Bialystok, Waszyngtona 13, 15-269 Białystok, Poland; (J.P.); (W.E.N.)
| | - Wiesława Ewa Niklińska
- Department of Histology and Embryology, Medical University in Bialystok, Waszyngtona 13, 15-269 Białystok, Poland; (J.P.); (W.E.N.)
| | - Adrian Chlanda
- Graphene and Composites Research Group, Łukasiewicz Research Network—Institute of Microelectronics and Photonics, al. Lotników 32/46, 02-668 Warszawa, Poland
| |
Collapse
|
23
|
Metastasis prevention: targeting causes and roots. Clin Exp Metastasis 2022; 39:505-519. [PMID: 35347574 DOI: 10.1007/s10585-022-10162-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022]
Abstract
The spread of tumor cells from the primary focus, metastasis, is the main cause of cancer mortality. Therefore, anticancer therapy should be focused on the prevention of metastatic disease. Key targets can be conditions in the primary tumor that are favorable for the appearance of metastatic cells and the first steps of the metastatic cascade. Here, we discuss different approaches for targeting metastasis causes (hypoxia, metabolism changes, and tumor microenvironment) and roots (angiogenesis, epithelial-mesenchymal transition, migration, and invasion). Also, we emphasize the challenges of the existing approaches for metastasis prevention and suggest opportunities to overcome them. In conclusion, we highlight the importance of clinical evaluation of the agents showing antimetastatic effects in vivo, especially in patients with early-stage cancers, the identification of metastatic seeds, and the development of therapeutics for their eradication.
Collapse
|
24
|
Ikeda-Imafuku M, Wang LLW, Rodrigues D, Shaha S, Zhao Z, Mitragotri S. Strategies to improve the EPR effect: A mechanistic perspective and clinical translation. J Control Release 2022; 345:512-536. [PMID: 35337939 DOI: 10.1016/j.jconrel.2022.03.043] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Many efforts have been made to achieve targeted delivery of anticancer drugs to enhance their efficacy and to reduce their adverse effects. These efforts include the development of nanomedicines as they can selectively penetrate through tumor blood vessels through the enhanced permeability and retention (EPR) effect. The EPR effect was first proposed by Maeda and co-workers in 1986, and since then various types of nanoparticles have been developed to take advantage of the phenomenon with regards to drug delivery. However, the EPR effect has been found to be highly variable and thus unreliable due to the complex tumor microenvironment. Various physical and pharmacological strategies have been explored to overcome this challenge. Here, we review key advances and emerging concepts of such EPR-enhancing strategies. Furthermore, we analyze 723 clinical trials of nanoparticles with EPR enhancers and discuss their clinical translation.
Collapse
Affiliation(s)
- Mayumi Ikeda-Imafuku
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Danika Rodrigues
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Suyog Shaha
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA
| | - Zongmin Zhao
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA; Translational Oncology Program, University of Illinois Cancer Center, Chicago, IL 60612, USA.
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Cambridge, MA 20138, USA.
| |
Collapse
|
25
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
26
|
Targeting nanoparticles to malignant tumors. Biochim Biophys Acta Rev Cancer 2022; 1877:188703. [DOI: 10.1016/j.bbcan.2022.188703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/01/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022]
|
27
|
Zhu D, Li Y, Zhang Z, Xue Z, Hua Z, Luo X, Zhao T, Lu C, Liu Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021; 19:435. [PMID: 34930293 PMCID: PMC8686559 DOI: 10.1186/s12951-021-01190-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor vessels can provide oxygen and nutrition for solid tumor tissue, create abnormal tumor microenvironment (TME), and play a vital role in the development, immune escape, metastasis and drug resistance of tumor. Tumor vessel-targeting therapy has become an important and promising direction in anti-tumor therapy, with the development of five anti-tumor therapeutic strategies, including vascular disruption, anti-angiogenesis, vascular blockade, vascular normalization and breaking immunosuppressive TME. However, the insufficient drug accumulation and severe side effects of vessel-targeting drugs limit their development in clinical application. Nanotechnology offers an excellent platform with flexible modified surface that can precisely deliver diverse cargoes, optimize efficacy, reduce side effects, and realize the combined therapy. Various nanomedicines (NMs) have been developed to target abnormal tumor vessels and specific TME to achieve more efficient vessel-targeting therapy. The article reviews tumor vascular abnormalities and the resulting abnormal microenvironment, the application of NMs in the tumor vessel-targeting strategies, and how NMs can improve these strategies and achieve multi-strategies combination to maximize anti-tumor effects. ![]()
Collapse
Affiliation(s)
- Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
28
|
Lahooti B, Poudel S, Mikelis CM, Mattheolabakis G. MiRNAs as Anti-Angiogenic Adjuvant Therapy in Cancer: Synopsis and Potential. Front Oncol 2021; 11:705634. [PMID: 34956857 PMCID: PMC8695604 DOI: 10.3389/fonc.2021.705634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a key mechanism for tumor growth and metastasis and has been a therapeutic target for anti-cancer treatments. Intensive vascular growth is concomitant with the rapidly proliferating tumor cell population and tumor outgrowth. Current angiogenesis inhibitors targeting either one or a few pro-angiogenic factors or a range of downstream signaling molecules provide clinical benefit, but not without significant side effects. miRNAs are important post-transcriptional regulators of gene expression, and their dysregulation has been associated with tumor progression, metastasis, resistance, and the promotion of tumor-induced angiogenesis. In this mini-review, we provide a brief overview of the current anti-angiogenic approaches, their molecular targets, and side effects, as well as discuss existing literature on the role of miRNAs in angiogenesis. As we highlight specific miRNAs, based on their activity on endothelial or cancer cells, we discuss their potential for anti-angiogenic targeting in cancer as adjuvant therapy and the importance of angiogenesis being evaluated in such combinatorial approaches.
Collapse
Affiliation(s)
- Behnaz Lahooti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
| | - Sagun Poudel
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, United States
| | - Constantinos M. Mikelis
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, United States
- Department of Pharmacy, University of Patras, Patras, Greece
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, United States
| |
Collapse
|
29
|
Biomembrane-based nanostructures for cancer targeting and therapy: From synthetic liposomes to natural biomembranes and membrane-vesicles. Adv Drug Deliv Rev 2021; 178:113974. [PMID: 34530015 DOI: 10.1016/j.addr.2021.113974] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/29/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
The translational success of liposomes in chemotherapeutics has already demonstrated the great potential of biomembrane-based nanostructure in effective drug delivery. Meanwhile, increasing efforts are being dedicated to the application of naturally derived lipid membranes, including cellular membranes and extracellular vesicles in anti-cancer therapies. While synthetic liposomes support superior multifunctional flexibility, natural biomembrane materials possess interesting biomimetic properties and can also be further engineered for intelligent design. Despite being remarkably different from each other in production and composition, the phospholipid bilayer structure in common allows liposomes, cell membrane-derived nanomaterials, and extracellular vesicles to be modified, functionalized, and exploited in many similar manners against challenges posed by tumor-targeted drug delivery. This review will summarize the recent advancements in engineering the membrane-derived nanostructures with "intelligent" modules to respond, regulate, and target tumor cells and the microenvironment to fight against malignancy. We will also discuss perspectives of combining engineered functionalities with naturally occurring activity for enhanced cancer therapy.
Collapse
|
30
|
Mathematical Analysis for the Effects of Medicine Supplies to a Solid Tumor. Symmetry (Basel) 2021. [DOI: 10.3390/sym13111988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objective: 1. Interpretation of the variations of solute medicine amount in blood vessels and TAF concentration with respect to the flow rates of injected drugs into liver and heart. 2. Description of the alteration of tumor cell density versus the time and radius variations. Methodology: Step 1. Compartmental analysis is adopted for the concentration of chemotaxis caused by injected substances L and H based on the assumption: two different medicines I1 and I2 are injected into heart and liver to recover the functions of each organ, respectively, without any side effects. Step 2. A partial differential equation is derived for the growth of TAF considering the diffusion of TAF and the rate of decay of TAF according to the disturbance of medicine M in blood vessels. Step 3. A partial differential equation is derived for the motion of tumor cells in the lights of random motility and chemotaxis in response to TAF gradients. Step 4. Exact solutions are obtained for the concentration of chemotaxis caused by injected substances L and H under the assumption that the loss of mass is proportional to mass itself. Step 5. Exact solution is obtained for the partial differential equation describing the growth of TAF using the separation of variables. Step 6. A finite volume approach is executed to search approximated solutions due to the complexity of the partial differential equation describing the motion of tumor cells. Results: 1. The concentration of medicine (M) decreases as the ratio of flow rate from heart into vessel to flow rate from liver into heart (k1k2) increases. 2. TAF concentration increases with the growth of the value of ratio k1k2 and TAF shows the smallest concentration when the flow rate of each injected medicine is similar. 3. Tumor cells react highly sensitive as soon as medicine supplies and tumor cell’s density is decreased drastically at the moment of medicine injection. 4. Tumor cell density decreases exponentially at an early stage and the density decrease is developed in a fluctuating manner along the radius. Conclusions: 1. The presented mathematical approach has the potential for the profound analysis of the variations of solute medicine amount in blood vessels, TAF concentration, and the alteration of tumor cell density according to the functional recoveries of liver and heart. 2. The mathematical approach may be applicable in the investigation of tumor cell’s behavior on the basis of complex interaction among five represented organs: kidney, liver, heart, spleen, and lung. A mathematical approach is developed to describe the variation of a solid tumor cell density in response to drug supply. The investigation is progressed based on the assumption that two different medicines, I1 and I2, are injected into heart and liver with flow rates k1 and k2 to recover the functions of each organ, respectively. A medicine function system for the reactions of tumor angiogenic factors (TAF) to medicine injection is obtained using a compartmental analysis. The mathematical governing equations for tumor cells motion are derived taking into account random motility and chemotaxis in response to TAF gradients and a finite volume method with time-changing is adopted to obtain numerical solutions due to the complexity of the governing equations. The variation of the flow rates k1 and k2 exerts profound influences on the concentration of medicine, and similar flow rate of k1 and k2 produces the greatest amount of medicine in blood vessels and suppresses strong inhibition in TAF movement. Tumor cells react very sensitively to drug injection and the tumor cell density decreases to less than 20% at an early stage of administration. However, the density of tumor cell diminishes slowly after the early stage of sudden change and the duration for complete therapy of tumor cells requires a long time.
Collapse
|
31
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
32
|
He JS, Liu SJ, Zhang YR, Chu XD, Lin ZB, Zhao Z, Qiu SH, Guo YG, Ding H, Pan YL, Pan JH. The Application of and Strategy for Gold Nanoparticles in Cancer Immunotherapy. Front Pharmacol 2021; 12:687399. [PMID: 34163367 PMCID: PMC8215714 DOI: 10.3389/fphar.2021.687399] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/24/2021] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy of malignant tumor is a verified and crucial anti-tumor strategy to help patients with cancer for prolonging prognostic survival. It is a novel anticancer tactics that activates the immune system to discern and damage cancer cells, thereby prevent them from proliferating. However, immunotherapy still faces many challenges in view of clinical efficacy and safety issues. Various nanomaterials, especially gold nanoparticles (AuNPs), have been developed not only for anticancer treatment but also for delivering antitumor drugs or combining other treatment strategies. Recently, some studies have focused on AuNPs for enhancing cancer immunotherapy. In this review, we summarized how AuNPs applicated as immune agents, drug carriers or combinations with other immunotherapies for anticancer treatment. AuNPs can not only act as immune regulators but also deliver immune drugs for cancer. Therefore, AuNPs are candidates for enhancing the efficiency and safety of cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yun-long Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jing-hua Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
33
|
Liang Q, Zhou L, Li Y, Liu J, Liu Y. Nano drug delivery system reconstruct tumour vasculature for the tumour vascular normalisation. J Drug Target 2021; 30:119-130. [PMID: 33960252 DOI: 10.1080/1061186x.2021.1927056] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The abnormal structure and function of blood vessels in the TME are obvious characteristics of the tumour. Abnormal blood vessels with high leakage support the occurrence of malignant tumours and increase the possibility of tumour cell invasion and metastasis. The formation of abnormal vascular also enhances immunosuppression and prevents the delivery of chemotherapy drugs to deeper tumours. Therefore, the normalisation of tumour blood vessels is a very promising approach to improve anti-tumour efficacy, aiming to restore the structural integrity of vessels and improve drug delivery efficiency and anti-tumour immunity. In this review, we have summarised strategies to improve cancer treatment that via nano drug delivery technology regulates the normalisation of tumour blood vessels. The treatment strategies related to the structure and function of tumour blood vessels such as angiogenesis factors, tumour-associated macrophages, tumour vascular endothelial cells, tumour-associated fibroblasts and immune checkpoints in the TME were mainly discussed. The normalisation of tumour blood vessels presents new opportunities and challenges for the more efficient delivery of nanoparticles to tumour tissues and cells and an innovative combination of treatments for cancer.
Collapse
Affiliation(s)
- Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Liyue Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yifan Li
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jinxia Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan, China.,Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
34
|
Trout CJ, Clapp JA, Griepenburg JC. Plasmonic carriers responsive to pulsed laser irradiation: a review of mechanisms, design, and applications. NEW J CHEM 2021. [DOI: 10.1039/d1nj02062e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review focuses on interactions which govern release from plasmonic carrier systems including liposomes, polymersomes, and nanodroplets under pulsed irradiation.
Collapse
Affiliation(s)
- Cory J. Trout
- Department of Physics, Rutgers University-Camden, 227 Penn Street, Camden, NJ 08102, USA
- Department of Applied Physics, Rutgers University-Newark, 101 Warren St., Newark, NJ 07102, USA
| | - Jamie A. Clapp
- Center for Computational and Integrative Biology, Rutgers University-Camden, NJ 08102, USA
| | - Julianne C. Griepenburg
- Department of Physics, Rutgers University-Camden, 227 Penn Street, Camden, NJ 08102, USA
- Center for Computational and Integrative Biology, Rutgers University-Camden, NJ 08102, USA
| |
Collapse
|
35
|
Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment. Bioact Mater 2020; 6:1973-1987. [PMID: 33426371 PMCID: PMC7773537 DOI: 10.1016/j.bioactmat.2020.12.010] [Citation(s) in RCA: 375] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor development and metastasis are closely related to the structure and function of the tumor microenvironment (TME). Recently, TME modulation strategies have attracted much attention in cancer immunotherapy. Despite the preliminary success of immunotherapeutic agents, their therapeutic effects have been restricted by the limited retention time of drugs in TME. Compared with traditional delivery systems, nanoparticles with unique physical properties and elaborate design can efficiently penetrate TME and specifically deliver to the major components in TME. In this review, we briefly introduce the substitutes of TME including dendritic cells, macrophages, fibroblasts, tumor vasculature, tumor-draining lymph nodes and hypoxic state, then review various nanoparticles targeting these components and their applications in tumor therapy. In addition, nanoparticles could be combined with other therapies, including chemotherapy, radiotherapy, and photodynamic therapy, however, the nanoplatform delivery system may not be effective in all types of tumors due to the heterogeneity of different tumors and individuals. The changes of TME at various stages during tumor development are required to be further elucidated so that more individualized nanoplatforms could be designed.
Collapse
Key Words
- AC-NPs, antigen-capturing nanoparticles
- ANG2, angiopoietin-2
- APCs, antigen-presenting cells
- Ab, antibodies
- Ag, antigen
- AuNCs, gold nanocages
- AuNPs, gold nanoparticles
- BBB, blood-brain barrier
- BTK, Bruton's tyrosine kinase
- Bcl-2, B-cell lymphoma 2
- CAFs, cancer associated fibroblasts
- CAP, cleavable amphiphilic peptide
- CAR-T, Chimeric antigen receptor-modified T-cell therapy
- CCL, chemoattractant chemokines ligand
- CTL, cytotoxic T lymphocytes
- CTLA4, cytotoxic lymphocyte antigen 4
- CaCO3, calcium carbonate
- Cancer immunotherapy
- DCs, dendritic cells
- DMMA, 2,3-dimethylmaleic anhydrid
- DMXAA, 5,6-dimethylxanthenone-4-acetic acid
- DSF/Cu, disulfiram/copper
- ECM, extracellular matrix
- EGFR, epidermal growth factor receptor
- EMT, epithelial-mesenchymal transition
- EPG, egg phosphatidylglycerol
- EPR, enhanced permeability and retention
- FAP, fibroblast activation protein
- FDA, the Food and Drug Administration
- HA, hyaluronic acid
- HB-GFs, heparin-binding growth factors
- HIF, hypoxia-inducible factor
- HPMA, N-(2-hydroxypropyl) methacrylamide
- HSA, human serum albumin
- Hypoxia
- IBR, Ibrutinib
- IFN-γ, interferon-γ
- IFP, interstitial fluid pressure
- IL, interleukin
- LMWH, low molecular weight heparin
- LPS, lipopolysaccharide
- M2NP, M2-like TAM dual-targeting nanoparticle
- MCMC, mannosylated carboxymethyl chitosan
- MDSCs, myeloid-derived suppressor cells
- MPs, microparticles
- MnO2, manganese dioxide
- NF-κB, nuclear factor κB
- NK, nature killer
- NO, nitric oxide
- NPs, nanoparticles
- Nanoparticles
- ODN, oligodeoxynucleotides
- PD-1, programmed cell death protein 1
- PDT, photodynamic therapy
- PFC, perfluorocarbon
- PHDs, prolyl hydroxylases
- PLGA, poly(lactic-co-glycolic acid)
- PS, photosensitizer
- PSCs, pancreatic stellate cells
- PTX, paclitaxel
- RBC, red-blood-cell
- RLX, relaxin-2
- ROS, reactive oxygen species
- SA, sialic acid
- SPARC, secreted protein acidic and rich in cysteine
- TAAs, tumor-associated antigens
- TAMs, tumor-associated macrophages
- TDPA, tumor-derived protein antigens
- TGF-β, transforming growth factor β
- TIE2, tyrosine kinase with immunoglobulin and epidermal growth factor homology domain 2
- TIM-3, T cell immunoglobulin domain and mucin domain-3
- TLR, Toll-like receptor
- TME, tumor microenvironment
- TNF-α, tumor necrosis factor alpha
- TfR, transferrin receptor
- Tregs, regulatory T cells
- Tumor microenvironment
- UPS-NP, ultra-pH-sensitive nanoparticle
- VDA, vasculature disrupting agent
- VEGF, vascular endothelial growth factor
- cDCs, conventional dendritic cells
- melittin-NP, melittin-lipid nanoparticle
- nMOFs, nanoscale metal-organic frameworks
- scFv, single-chain variable fragment
- siRNA, small interfering RNA
- tdLNs, tumor-draining lymph nodes
- α-SMA, alpha-smooth muscle actin
Collapse
|
36
|
Menzel L, Höpken UE, Rehm A. Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth. Front Immunol 2020; 11:591741. [PMID: 33343570 PMCID: PMC7744479 DOI: 10.3389/fimmu.2020.591741] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E. Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
37
|
Lan H, Zhang W, Jin K, Liu Y, Wang Z. Modulating barriers of tumor microenvironment through nanocarrier systems for improved cancer immunotherapy: a review of current status and future perspective. Drug Deliv 2020; 27:1248-1262. [PMID: 32865029 PMCID: PMC7470050 DOI: 10.1080/10717544.2020.1809559] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy suppresses and destroys tumors by re-activating and sustaining the tumor-immune process, and thus improving the immune response of the body to the tumor. Immunotherapeutic strategies are showing promising results in pre-clinical and clinical trials, however, tumor microenvironment (TME) is extremely immunosuppressive. Thus, their translation from labs to clinics still faces issues. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. The combination of nanotechnology with immunotherapy potentiates the effectiveness of immunotherapy by increasing delivery and retention, and by reducing immunomodulation toxicity. This review aims to highlight the barriers offered by TME for hindering the efficiency of immunotherapy for cancer treatment. Next, we highlight various nano-carriers based strategies for modulating those barriers for achieving better therapeutic efficacy of cancer immunotherapy with higher safety. This review will add to the body of scientific knowledge and will be a good reference material for academia and industries.
Collapse
Affiliation(s)
- Huanrong Lan
- Department of Breast and Thyroid Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Wei Zhang
- Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ketao Jin
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Yuyao Liu
- Department of Colorectal Surgery, Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang Province, China
| | - Zhen Wang
- Rehabilitation and Sports Medicine Research Institute of Zhejiang Province, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
38
|
Moradi Kashkooli F, Soltani M, Rezaeian M, Meaney C, Hamedi MH, Kohandel M. Effect of vascular normalization on drug delivery to different stages of tumor progression: In-silico analysis. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Fercoq F, Remion E, Vallarino-Lhermitte N, Alonso J, Raveendran L, Nixon C, Le Quesne J, Carlin LM, Martin C. Microfilaria-dependent thoracic pathology associated with eosinophilic and fibrotic polyps in filaria-infected rodents. Parasit Vectors 2020; 13:551. [PMID: 33160409 PMCID: PMC7648300 DOI: 10.1186/s13071-020-04428-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pulmonary manifestations are regularly reported in both human and animal filariasis. In human filariasis, the main known lung manifestations are the tropical pulmonary eosinophilia syndrome. Its duration and severity are correlated with the presence of microfilariae. Litomosoides sigmodontis is a filarial parasite residing in the pleural cavity of rodents. This model is widely used to understand the immune mechanisms that are established during infection and for the screening of therapeutic molecules. Some pulmonary manifestations during the patent phase of infection with L. sigmodontis have been described in different rodent hosts more or less permissive to infection. METHODS Here, the permissive Mongolian gerbil (Meriones unguiculatus) was infected with L. sigmodontis. Prevalence and density of microfilariae and adult parasites were evaluated. Lungs were analyzed for pathological signatures using immunohistochemistry and 3D imaging techniques (two-photon and light sheet microscopy). RESULTS Microfilaremia in gerbils was correlated with parasite load, as amicrofilaremic individuals had fewer parasites in their pleural cavities. Fibrotic polypoid structures were observed on both pleurae of infected gerbils. Polyps were of variable size and developed from the visceral mesothelium over the entire pleura. The larger polyps were vascularized and strongly infiltrated by immune cells such as eosinophils, macrophages or lymphocytes. The formation of these structures was induced by the presence of adult filariae since small and rare polyps were observed before patency, but they were exacerbated by the presence of gravid females and microfilariae. CONCLUSIONS Altogether, these data emphasize the role of host-specific factors in the pathogenesis of filarial infections.
Collapse
Affiliation(s)
- Frédéric Fercoq
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Estelle Remion
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France
| | - Nathaly Vallarino-Lhermitte
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France
| | - Joy Alonso
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France
| | - Lisy Raveendran
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France
| | - Colin Nixon
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - John Le Quesne
- Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| | - Leo M Carlin
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1GH, UK
| | - Coralie Martin
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM UMR 7245), Muséum national d'Histoire naturelle, CNRS, P52, 61 rue Buffon, 75005, Paris, France.
| |
Collapse
|
40
|
Manipulation of immune‒vascular crosstalk: new strategies towards cancer treatment. Acta Pharm Sin B 2020; 10:2018-2036. [PMID: 33304777 PMCID: PMC7714955 DOI: 10.1016/j.apsb.2020.09.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor vasculature is characterized by aberrant structure and function, resulting in immune suppressive profiles of tumor microenvironment through limiting immune cell infiltration into tumors, endogenous immune surveillance and immune cell function. Vascular normalization as a novel therapeutic strategy tends to prune some of the immature blood vessels and fortify the structure and function of the remaining vessels, thus improving immune stimulation and the efficacy of immunotherapy. Interestingly, the presence of "immune‒vascular crosstalk" enables the formation of a positive feedback loop between vascular normalization and immune reprogramming, providing the possibility to develop new cancer therapeutic strategies. The applications of nanomedicine in vascular-targeting therapy in cancer have gained increasing attention due to its specific physical and chemical properties. Here, we reviewed the recent advances of effective routes, especially nanomedicine, for normalizing tumor vasculature. We also summarized the development of enhancing nanoparticle-based anticancer drug delivery via the employment of transcytosis and mimicking immune cell extravasation. This review explores the potential to optimize nanomedicine-based therapeutic strategies as an alternative option for cancer treatment.
Collapse
|
41
|
Adityan S, Tran M, Bhavsar C, Wu SY. Nano-therapeutics for modulating the tumour microenvironment: Design, development, and clinical translation. J Control Release 2020; 327:512-532. [DOI: 10.1016/j.jconrel.2020.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
|
42
|
Joo JI, Choi M, Jang SH, Choi S, Park SM, Shin D, Cho KH. Realizing Cancer Precision Medicine by Integrating Systems Biology and Nanomaterial Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1906783. [PMID: 32253807 DOI: 10.1002/adma.201906783] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/19/2019] [Indexed: 06/11/2023]
Abstract
Many clinical trials for cancer precision medicine have yielded unsatisfactory results due to challenges such as drug resistance and low efficacy. Drug resistance is often caused by the complex compensatory regulation within the biomolecular network in a cancer cell. Recently, systems biological studies have modeled and simulated such complex networks to unravel the hidden mechanisms of drug resistance and identify promising new drug targets or combinatorial or sequential treatments for overcoming resistance to anticancer drugs. However, many of the identified targets or treatments present major difficulties for drug development and clinical application. Nanocarriers represent a path forward for developing therapies with these "undruggable" targets or those that require precise combinatorial or sequential application, for which conventional drug delivery mechanisms are unsuitable. Conversely, a challenge in nanomedicine has been low efficacy due to heterogeneity of cancers in patients. This problem can also be resolved through systems biological approaches by identifying personalized targets for individual patients or promoting the drug responses. Therefore, integration of systems biology and nanomaterial engineering will enable the clinical application of cancer precision medicine to overcome both drug resistance of conventional treatments and low efficacy of nanomedicine due to patient heterogeneity.
Collapse
Affiliation(s)
- Jae Il Joo
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Minsoo Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Seong-Hoon Jang
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sea Choi
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Sang-Min Park
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Dongkwan Shin
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| |
Collapse
|
43
|
Srivastava S, Zahra FT, Gupta N, Tullar PE, Srivastava SK, Mikelis CM. Low Dose of Penfluridol Inhibits VEGF-Induced Angiogenesis. Int J Mol Sci 2020; 21:755. [PMID: 31979394 PMCID: PMC7036977 DOI: 10.3390/ijms21030755] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 12/23/2022] Open
Abstract
: Metastasis is considered a major burden in cancer, being responsible for more than 90% of cancer-related deaths. Tumor angiogenesis is one of the main processes that lead to tumor metastasis. Penfluridol is a classic and commonly used antipsychotic drug, which has a great ability to cross the blood-brain barrier. Recent studies have revealed that penfluridol has significant anti-cancer activity in diverse tumors, such as metastatic breast cancer and glioblastoma. Here, we aim to identify the effect of low doses of penfluridol on tumor microenvironment and compare it with its effect on tumor cells. Although low concentration of penfluridol was not toxic for endothelial cells, it blocked angiogenesis in vitro and in vivo. In vitro, penfluridol inhibited VEGF-induced primary endothelial cell migration and tube formation, and in vivo, it blocked VEGF- and FGF-induced angiogenesis in the matrigel plug assay. VEGF-induced VEGFR2 phosphorylation and the downstream p38 and ERK signaling pathways were not affected in endothelial cells, although VEGF-induced Src and Akt activation were abrogated by penfluridol treatment. When cancer cells were treated with the same low concentration of penfluridol, basal Src activation levels were mildly impaired, thus impacting their cell migration and wound healing efficiency. The potential of cancer-induced paracrine effect on endothelial cells was explored, although that did not seem to be a player for angiogenesis. Overall, our data demonstrates that low penfluridol levels, similar to the ones clinically used for anti-psychotic conditions, suppress angiogenic efficiency in the tumor microenvironment.
Collapse
Affiliation(s)
- Suyash Srivastava
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA (F.T.Z.); (N.G.)
| | - Fatema Tuz Zahra
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA (F.T.Z.); (N.G.)
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Nehal Gupta
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA (F.T.Z.); (N.G.)
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Paul E. Tullar
- Department of Obstetrics and Gynecology, School of Medicine, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA;
| | - Sanjay K. Srivastava
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA (F.T.Z.); (N.G.)
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Constantinos M. Mikelis
- Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA (F.T.Z.); (N.G.)
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|