1
|
Wang J, Zhang Y, Chen H, Wu Y, Liu J, Che H, Zhang Y, Zhu X. Motor-Cargo Structured Nanotractors for Augmented NIR Phototherapy via Gas-Boosted Tumor Penetration and Respiration-Impaired Mitochondrial Dysfunction. Adv Healthc Mater 2024:e2402063. [PMID: 39380347 DOI: 10.1002/adhm.202402063] [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: 06/05/2024] [Revised: 09/26/2024] [Indexed: 10/10/2024]
Abstract
Tumor microenvironment, characterized by dense extracellular matrix and severe hypoxia, has caused pronounced resistance to photodynamic therapy (PDT). Herein, it has designed an artificial nitric oxide (NO) nanotractor with a unique "motor-cargo" structure, where a photoswitching upconversion nanoparticle (UCNP) core serves as the optical engine to harvest NIR light and asymmetrically coated mesoporous silica (SiO2) shell acts as a cargo unit to load nitric oxide (NO) fuel molecule (RBS, Roussin's black salt) and PDT photosensitizer (ZnPc, zinc phthalocyanine). Upon illumination by 980 nm light, the UCNP emits blue light to excite RBS salt and release NO gas. On one hand, NO is used as the driving force to propel the particle with a high speed of ≈194 µm s-1 that generates significant rupture stress (over 0.95 kPa) on cell membrane to promote cellular endocytosis and intratumoral penetration. On the other hand, NO enables to alleviate tumor hypoxia by inhibiting cellular respiration as an oxygen conserver. When the excitation is subsequently switched to 808 nm light, the UCNP emits red light, triggering ZnPc to produce large amount of reactive oxygen species for PDT treatment. This study explores Janus-typed nanostructures for cell-particle interaction and gas-assisted phototherapy, opening avenues for versatile bioapplications.
Collapse
Affiliation(s)
- Jing Wang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yi Zhang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Huadong Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yihan Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Jinliang Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Hailong Che
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yong Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Xiaohui Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
2
|
Zhang S, Wang H. Targeting the lung tumour stroma: harnessing nanoparticles for effective therapeutic interventions. J Drug Target 2024:1-27. [PMID: 39356091 DOI: 10.1080/1061186x.2024.2410462] [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: 07/29/2024] [Revised: 08/27/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Lung cancer remains an influential global health concern, necessitating the development of innovative therapeutic strategies. The tumour stroma, which is known as tumour microenvironment (TME) has a central impact on tumour expansion and treatment resistance. The stroma of lung tumours consists of numerous cells and molecules that shape an environment for tumour expansion. This environment not only protects tumoral cells against immune system attacks but also enables tumour stroma to attenuate the action of antitumor drugs. This stroma consists of stromal cells like cancer-associated fibroblasts (CAFs), suppressive immune cells, and cytotoxic immune cells. Additionally, the presence of stem cells, endothelial cells and pericytes can facilitate tumour volume expansion. Nanoparticles are hopeful tools for targeted drug delivery because of their extraordinary properties and their capacity to devastate biological obstacles. This review article provides a comprehensive overview of contemporary advancements in targeting the lung tumour stroma using nanoparticles. Various nanoparticle-based approaches, including passive and active targeting, and stimuli-responsive systems, highlighting their potential to improve drug delivery efficiency. Additionally, the role of nanotechnology in modulating the tumour stroma by targeting key components such as immune cells, extracellular matrix (ECM), hypoxia, and suppressive elements in the lung tumour stroma.
Collapse
Affiliation(s)
- Shushu Zhang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| | - Hui Wang
- Cancer Center (Oncology) Department, the Second Affiliated Hospital, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
3
|
Darguzyte M, Rama E, Rix A, Baier J, Hermann J, Rezvantalab S, Khedri M, Jankowski J, Kiessling F. Riboflavin-targeted polymers improve tolerance of paclitaxel while maintaining therapeutic efficacy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 58:102751. [PMID: 38705222 DOI: 10.1016/j.nano.2024.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/15/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Active targeting can enhance precision and efficacy of drug delivery systems (DDS) against cancers. Riboflavin (RF) is a promising ligand for active targeting due to its biocompatibility and high riboflavin-receptor expression in cancers. In this study, RF-targeted 4-arm polyethylene glycol (PEG) stars conjugated with Paclitaxel (PTX), named PEG PTX RF, were evaluated as a targeted DDS. In vitro, PEG PTX RF exhibited higher toxicity against tumor cells compared to the non-targeted counterpart (PEG PTX), while free PTX displayed the highest acute toxicity. In vivo, all treatments were similarly effective, but PEG PTX RF-treated tumors showed fewer proliferating cells, pointing to sustained therapy effects. Moreover, PTX-treated animals' body and liver weights were significantly reduced, whereas both remained stable in PEG PTX and PEG PTX RF-treated animals. Overall, our targeted and non-targeted DDS reduced PTX's adverse effects, with RF targeting promoted drug uptake in cancer cells for sustained therapeutic effect.
Collapse
Affiliation(s)
- Milita Darguzyte
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne, 50931 Cologne, Germany; Institute for Translational Immune-Oncology, Cancer Research Center Cologne-Essen (CCCE), University of Cologne, 50931 Cologne, Germany
| | - Elena Rama
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Jasmin Baier
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Juliane Hermann
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Sima Rezvantalab
- Department of Chemical Engineering, Urmia University of Technology, 57166-419, Urmia, Iran
| | - Mohammad Khedri
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN), 19839-63113 Tehran, Iran
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; Fraunhofer MEVIS, Institute for Medical Image Computing, Forckenbeckstrasse 55, 52074 Aachen, Germany.
| |
Collapse
|
4
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
5
|
Wang X, Liu X, Guo Y, Gong T, Lu W, Han M, Guo Y, Wang X. The Remarkable Anti-Breast Cancer Efficacy and Anti-Metastasis by Multifunctional Nanoparticles Co-Loading Squamocin, R848 and IR 780. Int J Nanomedicine 2024; 19:4679-4699. [PMID: 38803997 PMCID: PMC11129788 DOI: 10.2147/ijn.s448860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Background Breast cancer is a heterogeneous disease globally accounting for approximately 1 million new cases annually. Chemotherapy remains the main therapeutic option, but the antitumor efficacy needs to be improved. Methods Two multifunctional nanoparticles were developed in this paper using oleic acid and mPEG2k-PCL2k as the drug carriers. Squamocin (Squ) was employed as a chemotherapeutic agent. Resiquimod (R848) or ginsenoside Rh2 was co-encapsulated in the nanoparticles to remold the immunosuppressive tumor microenvironment, and IR780 was coloaded as a photosensitizer to realize photothermal therapy. Results The obtained Squ-R848-IR780 nanoparticles and Squ-Rh2-IR780 nanoparticles were uniformly spherical and approximately (162.200 ± 2.800) nm and (157.300 ± 1.1590) nm, respectively, in average diameter, with good encapsulation efficiency (above 85% for each drug), excellent stability in various physiological media and high photothermal conversion efficiency (24.10% and 22.58%, respectively). After intravenous administration, both nanoparticles quickly accumulated in the tumor and effectively enhanced the local temperature of the tumor to over 45 °C when irradiated by an 808 nm laser. At a low dose of 0.1 mg/kg, Squ nanoparticles treatment alone displayed a tumor inhibition rate of 55.28%, pulmonary metastasis inhibition rate of 59.47% and a mean survival time of 38 days, which were all higher than those of PTX injection (8 mg/kg) (43.64%, 25 days and 37.25%), indicating that Squ was a potent and effective antitumor agent. Both multifunctional nanoparticles, Squ-Rh2-IR780 nanoparticles and Squ-R848-IR780 nanoparticles, demonstrated even better therapeutic efficacy, with tumor inhibition rates of 90.02% and 97.28%, pulmonary metastasis inhibition rates of 95.42% and 98.09, and mean survival times of 46 days and 52 days, respectively. Conclusion The multifunctional nanoparticles coloaded with squamocin, R848 and IR 780 achieved extraordinary therapeutic efficacy and excellent antimetastasis activity and are thus promising in the future treatment of breast tumors and probably other tumors.
Collapse
Affiliation(s)
- Xi Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xinxin Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yaoyao Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Tingting Gong
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Wenmin Lu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, People’s Republic of China
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
6
|
Zhang H, Wang J, Wu R, Zheng B, Sang Y, Wang B, Song L, Hu Y, Ma X. Self-Supplied Reactive Oxygen Species-Responsive Mitoxantrone Polyprodrug for Chemosensitization-Enhanced Chemotherapy under Moderate Hyperthermia. Adv Healthc Mater 2024; 13:e2303631. [PMID: 38278138 DOI: 10.1002/adhm.202303631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/11/2023] [Indexed: 01/28/2024]
Abstract
Currently, the secondary development and modification of clinical drugs has become one of the research priorities. Researchers have developed a variety of TME-responsive nanomedicine carriers to solve certain clinical problems. Unfortunately, endogenous stimuli such as reactive oxygen species (ROS), as an important prerequisite for effective therapeutic efficacy, are not enough to achieve the expected drug release process, therefore, it is difficult to achieve a continuous and efficient treatment process. Herein, a self-supply ROS-responsive cascade polyprodrug (PMTO) is designed. The encapsulation of the chemotherapy drug mitoxantrone (MTO) in a polymer backbone could effectively reduce systemic toxicity when transported in vivo. After PMTO is degraded by endogenous ROS of the TME, another part of the polyprodrug backbone becomes cinnamaldehyde (CA), which can further enhance intracellular ROS, thereby achieving a sustained drug release process. Meanwhile, due to the disruption of the intracellular redox environment, the efficacy of chemotherapy drugs is enhanced. Finally, the anticancer treatment efficacy is further enhanced due to the mild hyperthermia effect of PMTO. In conclusion, the designed PMTO demonstrates remarkable antitumor efficacy, effectively addressing the limitations associated with MTO.
Collapse
Affiliation(s)
- Hongjie Zhang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Jing Wang
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Ruiying Wu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Benyan Zheng
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yanxiang Sang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Bibo Wang
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Lei Song
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Yuan Hu
- School of Chemistry and Materials Science, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, P. R. China
- State Key Laboratory of Fire Science, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230026, P. R. China
| | - Xiaopeng Ma
- The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| |
Collapse
|
7
|
Wang C, Xu J, Cheng X, Sun G, Li F, Nie G, Zhang Y. Anti-lymphangiogenesis for boosting drug accumulation in tumors. Signal Transduct Target Ther 2024; 9:89. [PMID: 38616190 PMCID: PMC11016544 DOI: 10.1038/s41392-024-01794-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 04/16/2024] Open
Abstract
The inadequate tumor accumulation of anti-cancer agents is a major shortcoming of current therapeutic drugs and remains an even more significant concern in the clinical prospects for nanomedicines. Various strategies aiming at regulating the intratumoral permeability of therapeutic drugs have been explored in preclinical studies, with a primary focus on vascular regulation and stromal reduction. However, these methods may trigger or facilitate tumor metastasis as a tradeoff. Therefore, there is an urgent need for innovative strategies that boost intratumoral drug accumulation without compromising treatment outcomes. As another important factor affecting drug tumor accumulation besides vasculature and stroma, the impact of tumor-associated lymphatic vessels (LVs) has not been widely considered. In the current research, we verified that anlotinib, a tyrosine kinase inhibitor with anti-lymphangiogenesis activity, and SAR131675, a selective VEGFR-3 inhibitor, effectively decreased the density of tumor lymphatic vessels in mouse cancer models, further enhancing drug accumulation in tumor tissue. By combining anlotinib with therapeutic drugs, including doxorubicin (Dox), liposomal doxorubicin (Lip-Dox), and anti-PD-L1 antibody, we observed improved anti-tumor efficacy in comparison with monotherapy regimens. Meanwhile, this strategy significantly reduced tumor metastasis and elicited stronger anti-tumor immune responses. Our work describes a new, clinically transferrable approach to augmenting intratumoral drug accumulation, which shows great potential to address the current, unsatisfactory efficacies of therapeutic drugs without introducing metastatic risk.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100190, China
- Sino-Danish Center for Education and Research, Beijing, 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xiaoyu Cheng
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ge Sun
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100190, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fenfen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China.
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100190, China.
- Sino-Danish Center for Education and Research, Beijing, 100190, China.
| | - Yinlong Zhang
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100190, China.
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
8
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
9
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
10
|
Agiba AM, Arreola-Ramírez JL, Carbajal V, Segura-Medina P. Light-Responsive and Dual-Targeting Liposomes: From Mechanisms to Targeting Strategies. Molecules 2024; 29:636. [PMID: 38338380 PMCID: PMC10856102 DOI: 10.3390/molecules29030636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
In recent years, nanocarriers have played an ever-increasing role in clinical and biomedical applications owing to their unique physicochemical properties and surface functionalities. Lately, much effort has been directed towards the development of smart, stimuli-responsive nanocarriers that are capable of releasing their cargos in response to specific stimuli. These intelligent-responsive nanocarriers can be further surface-functionalized so as to achieve active tumor targeting in a sequential manner, which can be simply modulated by the stimuli. By applying this methodological approach, these intelligent-responsive nanocarriers can be directed to different target-specific organs, tissues, or cells and exhibit on-demand controlled drug release that may enhance therapeutic effectiveness and reduce systemic toxicity. Light, an external stimulus, is one of the most promising triggers for use in nanomedicine to stimulate on-demand drug release from nanocarriers. Light-triggered drug release can be achieved through light irradiation at different wavelengths, either in the UV, visible, or even NIR region, depending on the photophysical properties of the photo-responsive molecule embedded in the nanocarrier system, the structural characteristics, and the material composition of the nanocarrier system. In this review, we highlighted the emerging functional role of light in nanocarriers, with an emphasis on light-responsive liposomes and dual-targeted stimuli-responsive liposomes. Moreover, we provided the most up-to-date photo-triggered targeting strategies and mechanisms of light-triggered drug release from liposomes and NIR-responsive nanocarriers. Lastly, we addressed the current challenges, advances, and future perspectives for the deployment of light-responsive liposomes in targeted drug delivery and therapy.
Collapse
Affiliation(s)
- Ahmed M. Agiba
- Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, Monterrey 64849, Mexico;
| | - José Luis Arreola-Ramírez
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
| | - Verónica Carbajal
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
| | - Patricia Segura-Medina
- Departamento de Investigación en Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Mexico City 14080, Mexico; (J.L.A.-R.); (V.C.)
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Mexico City 14380, Mexico
| |
Collapse
|
11
|
Schwartz-Duval A, Mackeyev Y, Mahmud I, Lorenzi PL, Gagea M, Krishnan S, Sokolov KV. Intratumoral Biosynthesis of Gold Nanoclusters by Pancreatic Cancer to Overcome Delivery Barriers to Radiosensitization. ACS NANO 2024; 18:1865-1881. [PMID: 38206058 PMCID: PMC10811688 DOI: 10.1021/acsnano.3c04260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024]
Abstract
Nanoparticle delivery to solid tumors is a prime challenge in nanomedicine. Here, we approach this challenge through the lens of biogeochemistry, the field that studies the flow of chemical elements within ecosystems as manipulated by living cellular organisms and their environments. We leverage biogeochemistry concepts related to gold cycling against pancreatic cancer, considering mammalian organisms as drivers for gold nanoparticle biosynthesis. Sequestration of gold nanoparticles within tumors has been demonstrated as an effective strategy to enhance radiotherapy; however, the desmoplasia of pancreatic cancer impedes nanoparticle delivery. Our strategy overcomes this barrier by applying an atomic-scale agent, ionic gold, for intratumoral gold nanoparticle biosynthesis. Our comprehensive studies showed the cancer-specific synthesis of gold nanoparticles from externally delivered gold ions in vitro and in a murine pancreatic cancer model in vivo; a substantial colocalization of gold nanoparticles (GNPs) with cancer cell nuclei in vitro and in vivo; a strong radiosensitization effect by the intracellularly synthesized GNPs; a uniform distribution of in situ synthesized GNPs throughout the tumor volume; a nearly 40-day total suppression of tumor growth in animal models of pancreatic cancer treated with a combination of gold ions and radiation that was also associated with a significantly higher median survival versus radiation alone (235 vs 102 days, respectively).
Collapse
Affiliation(s)
- Aaron
S. Schwartz-Duval
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yuri Mackeyev
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Iqbal Mahmud
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Philip L. Lorenzi
- Department
of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Mihai Gagea
- Department
of Veterinary Medicine & Surgery, The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| | - Sunil Krishnan
- Vivian
L. Smith Department of Neurosurgery, University
of Texas Health Science Center, Houston, Texas 77030, United States
| | - Konstantin V. Sokolov
- Department
of Imaging Physics, The University of Texas
MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
12
|
Liu Y, Corrales-Guerrero S, Kuo JC, Robb R, Nagy G, Cui T, Lee RJ, Williams TM. Improved Targeting and Safety of Doxorubicin through a Novel Albumin Binding Prodrug Approach. ACS OMEGA 2024; 9:977-987. [PMID: 38222540 PMCID: PMC10785662 DOI: 10.1021/acsomega.3c07163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024]
Abstract
Human serum albumin (HSA) improves the pharmacokinetic profile of drugs attached to it, making it an attractive carrier with proven clinical success. In our previous studies, we have shown that Caveolin-1 (Cav-1) and caveolae-mediated endocytosis play important roles in the uptake of HSA and albumin-bound drugs. Doxorubicin is an FDA-approved chemotherapeutic agent that is effective against multiple cancers, but its clinical applicability has been hampered by its high toxicity levels. In this study, a doxorubicin-prodrug was developed that could independently and avidly bind HSA in circulation, called IPBA-Dox. We first developed and characterized IPBA-Dox and confirmed that it can bind albumin in vitro while retaining a potent cytotoxic effect. We then verified that it efficiently binds to HSA in circulation, leading to an improvement in the pharmacokinetic profile of the drug. In addition, we tested our prodrug for Cav-1 selectivity and found that it preferentially affects cells that express relatively higher levels of Cav-1 in vitro and in vivo. Moreover, we found that our compound was well tolerated in vivo at concentrations at which doxorubicin was lethal. Altogether, we have developed a doxorubicin-prodrug that can successfully bind HSA, retaining a strong cytotoxic effect that preferentially targets Cav-1 positive cells while improving the general tolerability of the drug.
Collapse
Affiliation(s)
- Yang Liu
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Sergio Corrales-Guerrero
- Biomedical
Sciences Graduate Program, The Ohio State
University, Columbus, Ohio 43210-1132, United States
| | - Jimmy C. Kuo
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Ryan Robb
- University
of North Carolina, Chapel
Hill, North Carolina 27514-3916, United States
| | - Gregory Nagy
- Biomedical
Sciences Graduate Program, The Ohio State
University, Columbus, Ohio 43210-1132, United States
| | - Tiantian Cui
- Department
of Radiation Oncology, City of Hope National
Medical Center, Duarte, California 91010, United States
| | - Robert J. Lee
- Division
of Pharmaceutics and Pharmacology, The Ohio
State University, Columbus, Ohio 43210-1132, United States
| | - Terence M. Williams
- Department
of Radiation Oncology, City of Hope National
Medical Center, Duarte, California 91010, United States
| |
Collapse
|
13
|
McWhorter R, Bonavida B. The Role of TAMs in the Regulation of Tumor Cell Resistance to Chemotherapy. Crit Rev Oncog 2024; 29:97-125. [PMID: 38989740 DOI: 10.1615/critrevoncog.2024053667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Tumor-associated macrophages (TAMs) are the predominant cell infiltrate in the immunosuppressive tumor microenvironment (TME). TAMs are central to fostering pro-inflammatory conditions, tumor growth, metastasis, and inhibiting therapy responses. Many cancer patients are innately refractory to chemotherapy and or develop resistance following initial treatments. There is a clinical correlation between the level of TAMs in the TME and chemoresistance. Hence, the pivotal role of TAMs in contributing to chemoresistance has garnered significant attention toward targeting TAMs to reverse this resistance. A prerequisite for such an approach requires a thorough understanding of the various underlying mechanisms by which TAMs inhibit response to chemotherapeutic drugs. Such mechanisms include enhancing drug efflux, regulating drug metabolism and detoxification, supporting cancer stem cell (CSCs) resistance, promoting epithelial-mesenchymal transition (EMT), inhibiting drug penetration and its metabolism, stimulating angiogenesis, impacting inhibitory STAT3/NF-κB survival pathways, and releasing specific inhibitory cytokines including TGF-β and IL-10. Accordingly, several strategies have been developed to overcome TAM-modulated chemoresistance. These include novel therapies that aim to deplete TAMs, repolarize them toward the anti-tumor M1-like phenotype, or block recruitment of monocytes into the TME. Current results from TAM-targeted treatments have been unimpressive; however, the use of TAM-targeted therapies in combination appears promising These include targeting TAMs with radiotherapy, chemotherapy, chemokine receptor inhibitors, immunotherapy, and loaded nanoparticles. The clinical limitations of these strategies are discussed.
Collapse
Affiliation(s)
| | - Benjamin Bonavida
- Department of Microbiology, Immunology, & Molecular Genetics, David Geffen School of Medicine at UCLA, Johnson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA 90025-1747, USA
| |
Collapse
|
14
|
Wu C, Sun Q, Liu X, Sun X, Chen Z, Shan H. Indocyanine Green-Loaded Liposomes-Assisted Photoacoustic Computed Tomography for Evaluating In Vivo Tumor Penetration of Liposomal Nanocarriers. MICROMACHINES 2023; 15:90. [PMID: 38258209 PMCID: PMC10820658 DOI: 10.3390/mi15010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/11/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024]
Abstract
Liposomes possess the potential to enhance drug solubility, prolong the duration of circulation, and augment drug accumulation at the tumor site through passive and active targeting strategies. However, there is a lack of studies examining the in vivo tumor penetration capabilities of liposomes of varying sizes, which hampers the development of drug delivery systems utilizing liposomal nanocarriers. Here, we present an indocyanine green (ICG)-loaded liposomes-assisted photoacoustic computed tomography (PACT) for directly evaluating the tumor penetration ability of liposomal nanocarriers in vivo. Through the utilization of microfluidic mixing combined with extrusion techniques, we successfully prepare liposomes encapsulating ICG in both large (192.6 ± 8.0 nm) and small (61.9 ± 0.6 nm) sizes. Subsequently, we designed a dual-wavelength PACT system to directly monitor the in vivo tumor penetration of large- and small-size ICG-encapsulated liposomes. In vivo PACT experiments indicate that ICG-loaded liposomes of smaller size exhibit enhanced penetration capability within tumor tissues. Our work presents a valuable approach to directly assess the penetration ability of liposomal nanocarriers in vivo, thereby facilitating the advancement of drug delivery systems with enhanced tumor penetration and therapeutic efficacy.
Collapse
Affiliation(s)
- Chenjun Wu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
- Changjun Riverside Middle School, Changsha 410023, China
| | - Qi Sun
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xiangdong Liu
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Xin Sun
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Zeyu Chen
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Han Shan
- State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
15
|
Cooley MB, Wegierak D, Perera R, Abenojar EC, Nittayacharn PA, Berg FM, Kim Y, Kolios MC, Exner AA. Assessing Tumor Microenvironment Characteristics and Stratifying EPR with a Nanobubble Companion Nanoparticle via Contrast-Enhanced Ultrasound Imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567934. [PMID: 38045236 PMCID: PMC10690218 DOI: 10.1101/2023.11.20.567934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The tumor microenvironment is characterized by dysfunctional endothelial cells, resulting in heightened vascular permeability. Many nanoparticle-based drug delivery systems attempt to use this enhanced permeability combined with impaired lymphatic drainage (a concept known as the 'enhanced permeability and retention effect' or EPR effect) as the primary strategy for drug delivery, but this has not proven to be as clinically effective as anticipated. The specific mechanisms behind the inconsistent clinical outcomes of nanotherapeutics have not been clearly articulated, and the field has been hampered by a lack of accessible tools to study EPR-associated phenomena in clinically relevant scenarios. While medical imaging has tremendous potential to contribute to this area, it has not been broadly explored. This work examines, for the first time, the use of multiparametric dynamic contrast-enhanced ultrasound (CEUS) with a novel nanoscale contrast agent to examine tumor microenvironment characteristics noninvasively and in real-time. We demonstrate that CEUS imaging can: (1) evaluate tumor microenvironment features and (2) be used to help predict the distribution of doxorubicin-loaded liposomes in the tumor parenchyma. CEUS using nanobubbles (NBs) was carried out in two tumor types of high (LS174T) and low (U87) vascular permeability, and time-intensity curve (TIC) parameters were evaluated in both models prior to injection of doxorubicin liposomes. Consistently, LS174T tumors showed significantly different TIC parameters, including area under the rising curve (2.7x), time to peak intensity (1.9x) and decorrelation time (DT, 1.9x) compared to U87 tumors. Importantly, the DT parameter successfully predicted tumoral nanoparticle distribution (r = 0.86 ± 0.13). Ultimately, substantial differences in NB-CEUS generated parameters between LS174T and U87 tumors suggest that this method may be useful in determining tumor vascular permeability and could be used as a biomarker for identifying tumor characteristics and predicting sensitivity to nanoparticle-based therapies. These findings could ultimately be applied to predicting treatment efficacy and to evaluating EPR in other diseases with pathologically permeable vasculature.
Collapse
|
16
|
Biancacci I, De Santis D, Rama E, Benderski K, Momoh J, Pohlberger R, Moeckel D, Kaps L, Rijcken CJF, Prakash J, Thewissen M, Kiessling F, Shi Y, Peña Q, Sofias AM, Consolino L, Lammers T. Repurposing Tamoxifen for Tumor Microenvironment Priming and Enhanced Tumor-Targeted Drug Delivery. ADVANCED THERAPEUTICS 2023; 6:adtp.202300098. [PMID: 39376929 PMCID: PMC7616667 DOI: 10.1002/adtp.202300098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Indexed: 10/09/2024]
Abstract
The dense stromal matrix in fibrotic tumors hinders tumor-targeted drug delivery. Tamoxifen (TMX), an estrogen receptor modulator that is clinically used for the treatment of breast cancer, has been shown to reprogram the tumor microenvironment (TME) and to alleviate desmoplasia. We here investigated if TMX, administered in free and nano-formulated form, can be repurposed as a TME remodeling agent to improve tumor accumulation of nano-formulations in pancreatic ductal adenocarcinoma and triple-negative breast cancer mouse models, evaluated using clinical-stage Cy7-labeled core-crosslinked polymeric micelles (CCPM). Under control conditions, we found higher levels of Cy7-CCPM in PANC-1 tumors (16.7 % ID g-1 at 48 h post i.v. injection) than in 4T1 tumors (11.0 % ID g-1). In both models, free and nano-formulated TMX failed to improve CCPM delivery. These findings were congruent with the results from histopathological immunofluorescence analysis of tumor tissue, which indicated that TMX treatment did not significantly change vascularization, perfusion, macrophage infiltration, collagen density, and collagen fiber thickness. Altogether, our results demonstrate that in PANC-1 and 4T1 mouse models, TMX treatment does not contribute to beneficial TME priming and enhanced tumor-targeted drug delivery.
Collapse
Affiliation(s)
- Ilaria Biancacci
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Daniele De Santis
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
- University of Urbino, Department of Biomolecular Sciences, Piazza Rinascimento 6, 61029, Urbino, Italy
| | - Elena Rama
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Karina Benderski
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Jeffrey Momoh
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Robert Pohlberger
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Diana Moeckel
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Leonard Kaps
- University Medical Center of the Johannes Gutenberg-University Mainz, Department of Internal Medicine 1, Langenbeckstrasse 1, 55131, Mainz, Germany
- University Medical Center of the Johannes Gutenberg-University Mainz, Institute of Translational Immunology and Research Center for Immunotherapy/FZI), Obere Zahlbacher Str. 63, 55131, Mainz, Germany
| | | | - Jai Prakash
- University of Twente, Engineered Therapeutics Section, Department of Advanced Organ Bioengineering and Therapeutics, Technical Medical Centre, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Marielle Thewissen
- Cristal Therapeutics, Oxfordlaan 55, 6229 EV, Maastricht, The Netherlands
| | - Fabian Kiessling
- RWTH Aachen University, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Max-Von-Laue-Strasse 2, 28359, Bremen, Germany
| | - Yang Shi
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Quim Peña
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Alexandros Marios Sofias
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Lorena Consolino
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| | - Twan Lammers
- RWTH Aachen University, Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
17
|
Wang C, Xu J, Zhang Y, Nie G. Emerging nanotechnological approaches to regulating tumor vasculature for cancer therapy. J Control Release 2023; 362:647-666. [PMID: 37703928 DOI: 10.1016/j.jconrel.2023.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Abnormal angiogenesis stands for one of the most striking manifestations of malignant tumor. The pathologically and structurally abnormal tumor vasculature facilitates a hostile tumor microenvironment, providing an ideal refuge exclusively for cancer cells. The emergence of vascular regulation drugs has introduced a distinctive class of therapeutics capable of influencing nutrition supply and drug delivery efficacy without the need to penetrate a series of physical barriers to reach tumor cells. Nanomedicines have been further developed for more precise regulation of tumor vasculature with the capacity of co-delivering multiple active pharmaceutical ingredients, which overall reduces the systemic toxicity and boosts the therapeutic efficacy of free drugs. Additionally, precise structure design enables the integration of specific functional motifs, such as surface-targeting ligands, droppable shells, degradable framework, or stimuli-responsive components into nanomedicines, which can improve tissue-specific accumulation, enhance tissue penetration, and realize the controlled and stimulus-triggered release of the loaded cargo. This review describes the morphological and functional characteristics of tumor blood vessels and summarizes the pivotal molecular targets commonly used in nanomedicine design, and then highlights the recent cutting-edge advancements utilizing nanotechnologies for precise regulation of tumor vasculature. Finally, the challenges and future directions of this field are discussed.
Collapse
Affiliation(s)
- Chunling Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yinlong Zhang
- Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; School of Nanoscience and Engineering, School of Chemical Science, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China; Sino-Danish Center for Education and Research, Sino-Danish College of UCAS, Beijing 100190, China; GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China.
| |
Collapse
|
18
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
19
|
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: 2] [Impact Index Per Article: 2.0] [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
|
20
|
Wang Q, Yang H, Liu Y, Zhou Z, Zhang X, Sang M, Xu F, Song L, Xia T, Zhang Y, Wei J, Zhang X, Ding Q. Awakening Allies for Breaking Microenvironment Barriers: NIR-II Guided Orthogonal Activation of Tumor-Infiltrating Mast Cells for Efficient Nano-Drug Delivery. Adv Healthc Mater 2023; 12:e2300420. [PMID: 37141500 DOI: 10.1002/adhm.202300420] [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: 02/09/2023] [Revised: 04/29/2023] [Indexed: 05/06/2023]
Abstract
Mast cells (MCs), powerful immune cells that heavily infiltrate cancer cells, play a crucial role in tumor formation. Activated MCs can release histamine and a family of proteases through degranulation effects, concurrently achieving endothelial junction weakening and stromal degradation of the tumor microenvironment, thereby clearing the obstacles for nano-drug infiltration. To achieve precise activation of tumor-infiltrating MCs, orthogonally excited rare earth nanoparticles (ORENP), with two channels, are introduced for the controllable stimulating drugs release wrapped in "photocut tape". The ORENP can emit near-infrared II (NIR-II) for image tracing for tumor localization in Channel 1 (808/NIR-II) and allows energy upconversion to emit ultraviolet (UV) light for releasing drugs for MCs stimulation in Channel 2 (980/UV). Finally, the combined use of chemical and cellular tools enables clinical nano-drugs to achieve a significant increase in tumor infiltration, thereby enhancing the efficacy of nano-chemotherapy.
Collapse
Affiliation(s)
- Qingyuan Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Haiyan Yang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yuan Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Zhaoxi Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xu Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Mingyi Sang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Feng Xu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Lebin Song
- Department of Dermatology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Yue Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Jifu Wei
- Department of Pharmacy, Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing, 210009, China
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing, 211103, China
| | - Xiaobo Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Qiang Ding
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| |
Collapse
|
21
|
Ottaiano A, Ianniello M, Santorsola M, Ruggiero R, Sirica R, Sabbatino F, Perri F, Cascella M, Di Marzo M, Berretta M, Caraglia M, Nasti G, Savarese G. From Chaos to Opportunity: Decoding Cancer Heterogeneity for Enhanced Treatment Strategies. BIOLOGY 2023; 12:1183. [PMID: 37759584 PMCID: PMC10525472 DOI: 10.3390/biology12091183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023]
Abstract
Cancer manifests as a multifaceted disease, characterized by aberrant cellular proliferation, survival, migration, and invasion. Tumors exhibit variances across diverse dimensions, encompassing genetic, epigenetic, and transcriptional realms. This heterogeneity poses significant challenges in prognosis and treatment, affording tumors advantages through an increased propensity to accumulate mutations linked to immune system evasion and drug resistance. In this review, we offer insights into tumor heterogeneity as a crucial characteristic of cancer, exploring the difficulties associated with measuring and quantifying such heterogeneity from clinical and biological perspectives. By emphasizing the critical nature of understanding tumor heterogeneity, this work contributes to raising awareness about the importance of developing effective cancer therapies that target this distinct and elusive trait of cancer.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Monica Ianniello
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (M.I.); (R.R.); (R.S.); (G.S.)
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Raffaella Ruggiero
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (M.I.); (R.R.); (R.S.); (G.S.)
| | - Roberto Sirica
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (M.I.); (R.R.); (R.S.); (G.S.)
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy;
| | - Francesco Perri
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Marco Cascella
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Massimiliano Di Marzo
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via Luigi De Crecchio 7, 80138 Naples, Italy;
| | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (F.P.); (M.C.); (M.D.M.); (G.N.)
| | - Giovanni Savarese
- AMES, Centro Polidiagnostico Strumentale srl, Via Padre Carmine Fico 24, 80013 Casalnuovo Di Napoli, Italy; (M.I.); (R.R.); (R.S.); (G.S.)
| |
Collapse
|
22
|
Desai P, Dasgupta A, Sofias AM, Peña Q, Göstl R, Slabu I, Schwaneberg U, Stiehl T, Wagner W, Jockenhövel S, Stingl J, Kramann R, Trautwein C, Brümmendorf TH, Kiessling F, Herrmann A, Lammers T. Transformative Materials for Interfacial Drug Delivery. Adv Healthc Mater 2023; 12:e2301062. [PMID: 37282805 PMCID: PMC11468550 DOI: 10.1002/adhm.202301062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/25/2023] [Indexed: 06/08/2023]
Abstract
Drug delivery systems (DDS) are designed to temporally and spatially control drug availability and activity. They assist in improving the balance between on-target therapeutic efficacy and off-target toxic side effects. DDS aid in overcoming biological barriers encountered by drug molecules upon applying them via various routes of administration. They are furthermore increasingly explored for modulating the interface between implanted (bio)medical materials and host tissue. Herein, an overview of the biological barriers and host-material interfaces encountered by DDS upon oral, intravenous, and local administration is provided, and material engineering advances at different time and space scales to exemplify how current and future DDS can contribute to improved disease treatment are highlighted.
Collapse
Affiliation(s)
- Prachi Desai
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
| | - Quim Peña
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Robert Göstl
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
| | - Ioana Slabu
- Institute of Applied Medical EngineeringHelmholtz InstituteMedical FacultyRWTH Aachen University52074AachenGermany
| | | | - Thomas Stiehl
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Institute for Computational Biomedicine – Disease ModelingRWTH Aachen University52074AachenGermany
| | - Wolfgang Wagner
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
- Institute for Stem Cell BiologyUniversity Hospital of RWTH Aachen52074AachenGermany
| | - Stefan Jockenhövel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz Institute AachenRWTH Aachen University52074AachenGermany
| | - Julia Stingl
- Institute of Clinical PharmacologyUniversity Hospital RWTH Aachen52074AachenGermany
| | - Rafael Kramann
- Division of Nephrology and Clinical ImmunologyRWTH Aachen University52074AachenGermany
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University52074AachenGermany
| | - Christian Trautwein
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of Medicine III (GastroenterologyMetabolic diseases and Intensive Care)University Hospital RWTH Aachen52074AachenGermany
| | - Tim H. Brümmendorf
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of HematologyOncologyHemostaseology and Stem Cell TransplantationRWTH Aachen University Medical School52074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| | - Andreas Herrmann
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
- Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 152074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| |
Collapse
|
23
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
24
|
Yao H, Xu H, Wu M, Lei W, Li L, Liu D, Wang Z, Ran H, Ma H, Zhou X. Targeted long-term noninvasive treatment of choroidal neovascularization by biodegradable nanoparticles. Acta Biomater 2023; 166:536-551. [PMID: 37196903 DOI: 10.1016/j.actbio.2023.05.021] [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: 11/04/2022] [Revised: 04/24/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
Choroidal neovascularization (CNV) is the main cause of vision loss in patients with wet age-related macular degeneration (AMD). Currently, treatment of these conditions requires repeated intravitreal injections, which may lead to complications such as infection and hemorrhage. So, we have developed a noninvasive method for treating CNV with nanoparticles, namely, Angiopoietin1-anti CD105-PLGA nanoparticles (AAP NPs), which targets the CNV to enhance drug accumulation at the site. These nanoparticles, with PLGA as a carrier, can slowly release encapsulated Angiopoietin 1 (Ang 1) and target the choroidal neovascularization marker CD105 to enhance drug accumulation, increases vascular endothelial cadherin (VE-cadherin) expression between vascular endothelial cells, effectively reduce neovascularization leakage and inhibit Angiopoietin 2(Ang 2) secretion by endothelial cells. In a rat model of laser-induced CNV, intravenous injection of AAP NPs exerted a good therapeutic effect in reducing CNV leakage and area. In short, these synthetic AAP NPs provide an effective alternative treatment for AMD and meet the urgent need for noninvasive treatment in neovascular ophthalmopathy. STATEMENT OF SIGNIFICANCE: This work describes the synthesis, injection-mediated delivery, in vitro and in vivo efficacy of targeted nanoparticles with encapsulated Ang1; via these nanoparticles, the drug can be targeted to choroidal neovascularization lesions for continuous treatment. The release of Ang1 can effectively reduce neovascularization leakage, maintain vascular stability, and inhibit Ang2 secretion and inflammation. This study provides a new approach for the treatment of wet age-related macular degeneration.
Collapse
Affiliation(s)
- Hao Yao
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Huan Xu
- Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Mingxing Wu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China
| | - Wulong Lei
- Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lanjiao Li
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Danning Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Huafeng Ma
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China.
| | - Xiyuan Zhou
- Department of Ophthalmology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400012, China; Chongqing Key Laboratory of Ophthalmology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China.
| |
Collapse
|
25
|
Onishchenko NR, Moskovtsev AA, Kobanenko MK, Tretiakova DS, Alekseeva AS, Kolesov DV, Mikryukova AA, Boldyrev IA, Kapkaeva MR, Shcheglovitova ON, Bovin NV, Kubatiev AA, Tikhonova OV, Vodovozova EL. Protein Corona Attenuates the Targeting of Antitumor Sialyl Lewis X-Decorated Liposomes to Vascular Endothelial Cells under Flow Conditions. Pharmaceutics 2023; 15:1754. [PMID: 37376203 DOI: 10.3390/pharmaceutics15061754] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Previously, we showed in the human umbilical vein endothelial cells (HUVECs) model that a liposome formulation of melphalan lipophilic prodrug (MlphDG) decorated with selectin ligand tetrasaccharide Sialyl Lewis X (SiaLeX) undergoes specific uptake by activated cells and in an in vivo tumor model causes a severe antivascular effect. Here, we cultured HUVECs in a microfluidic chip and then applied the liposome formulations to study their interactions with the cells in situ under hydrodynamic conditions close to capillary blood flow using confocal fluorescent microscopy. The incorporation of 5 to 10% SiaLeX conjugate in the bilayer of MlphDG liposomes increased their consumption exclusively by activated endotheliocytes. The increase of serum concentration from 20 to 100% in the flow resulted in lower liposome uptake by the cells. To elucidate the possible roles of plasma proteins in the liposome-cell interactions, liposome protein coronas were isolated and analyzed by shotgun proteomics and immunoblotting of selected proteins. Proteomic analysis showed that a gradual increase in SiaLeX content correlated with the overall enrichment of the liposome-associated proteins with several apolipoproteins, including the most positively charged one, ApoC1, and serum amyloid A4, associated with inflammation, on the one hand, and a decrease in the content of bound immunoglobulins, on the other. The article discusses the potential interference of the proteins in the binding of liposomes to selectins of endothelial cells.
Collapse
Affiliation(s)
- Natalia R Onishchenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Alexey A Moskovtsev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Maria K Kobanenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Daria S Tretiakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Anna S Alekseeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Dmitry V Kolesov
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Anna A Mikryukova
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Ivan A Boldyrev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Marina R Kapkaeva
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, ul. Gamaleya 18, 123098 Moscow, Russia
| | - Olga N Shcheglovitova
- N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Healthcare of the Russian Federation, ul. Gamaleya 18, 123098 Moscow, Russia
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| | - Aslan A Kubatiev
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, ul. Baltiyskaya 8, 125315 Moscow, Russia
| | - Olga V Tikhonova
- Institute of Biomedical Chemistry, ul. Pogodinskaya 10, 119121 Moscow, Russia
| | - Elena L Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997 Moscow, Russia
| |
Collapse
|
26
|
Honari A, Sirsi SR. The Evolution and Recent Trends in Acoustic Targeting of Encapsulated Drugs to Solid Tumors: Strategies beyond Sonoporation. Pharmaceutics 2023; 15:1705. [PMID: 37376152 DOI: 10.3390/pharmaceutics15061705] [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: 04/13/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Despite recent advancements in ultrasound-mediated drug delivery and the remarkable success observed in pre-clinical studies, no delivery platform utilizing ultrasound contrast agents has yet received FDA approval. The sonoporation effect was a game-changing discovery with a promising future in clinical settings. Various clinical trials are underway to assess sonoporation's efficacy in treating solid tumors; however, there are disagreements on its applicability to the broader population due to long-term safety issues. In this review, we first discuss how acoustic targeting of drugs gained importance in cancer pharmaceutics. Then, we discuss ultrasound-targeting strategies that have been less explored yet hold a promising future. We aim to shed light on recent innovations in ultrasound-based drug delivery including newer designs of ultrasound-sensitive particles specifically tailored for pharmaceutical usage.
Collapse
Affiliation(s)
- Arvin Honari
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shashank R Sirsi
- Department of Bioengineering, Erik Johnson School of Engineering, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
27
|
Lee J, Choi MK, Song IS. Recent Advances in Doxorubicin Formulation to Enhance Pharmacokinetics and Tumor Targeting. Pharmaceuticals (Basel) 2023; 16:802. [PMID: 37375753 PMCID: PMC10301446 DOI: 10.3390/ph16060802] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX), a widely used drug in cancer chemotherapy, induces cell death via multiple intracellular interactions, generating reactive oxygen species and DNA-adducted configurations that induce apoptosis, topoisomerase II inhibition, and histone eviction. Despite its wide therapeutic efficacy in solid tumors, DOX often induces drug resistance and cardiotoxicity. It shows limited intestinal absorption because of low paracellular permeability and P-glycoprotein (P-gp)-mediated efflux. We reviewed various parenteral DOX formulations, such as liposomes, polymeric micelles, polymeric nanoparticles, and polymer-drug conjugates, under clinical use or trials to increase its therapeutic efficacy. To improve the bioavailability of DOX in intravenous and oral cancer treatment, studies have proposed a pH- or redox-sensitive and receptor-targeted system for overcoming DOX resistance and increasing therapeutic efficacy without causing DOX-induced toxicity. Multifunctional formulations of DOX with mucoadhesiveness and increased intestinal permeability through tight-junction modulation and P-gp inhibition have also been used as orally bioavailable DOX in the preclinical stage. The increasing trends of developing oral formulations from intravenous formulations, the application of mucoadhesive technology, permeation-enhancing technology, and pharmacokinetic modulation with functional excipients might facilitate the further development of oral DOX.
Collapse
Affiliation(s)
- Jihoon Lee
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheon-an 31116, Republic of Korea;
| | - Im-Sook Song
- BK21 FOUR Community-Based Intelligent Novel Drug Discovery Education Unit, Vessel-Organ Interaction Research Center (VOICE), Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
28
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
29
|
Xie Y, Ren Z, Chen H, Tang H, Zhu M, Lv Z, Bao H, Zhang Y, Liu R, Shen Y, Zheng Y, Miao D, Guo X, Chen H, Wang S, Pei J. A novel estrogen-targeted PEGylated liposome co-delivery oxaliplatin and paclitaxel for the treatment of ovarian cancer. Biomed Pharmacother 2023; 160:114304. [PMID: 36724638 DOI: 10.1016/j.biopha.2023.114304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/14/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Ovarian cancer is the second cause of death among gynecological malignancies. In this study, we designed a novel estrogen-targeted PEGylated liposome loaded with oxaliplatin and paclitaxel (ES-SSL-OXA/PTX) which could target estrogen receptor (ER) highly expressed on the surface of SKOV-3 cells to enhance therapeutic efficacy and reduce the side effects for SKOV-3 tumor therapy. ES-SSL-OXA/PTX was prepared by thin film hydration method and exhibited a uniform spherical morphology. Encapsulation efficiency (EE) were determined by HPLC method with the results of 44.10% for OXA and 65.85% for PTX. The mean particle size and polydispersity index (PDI) were 168.46 nm and 0.145, respectively. In vivo and in vitro targeting study confirmed that ES-SSL-OXA/PTX has optimum specific targeting ability. Meanwhile, In vitro and in vivo antitumor results of ES-SSL-OXA/PTX exhibited a superior antiproliferative effect on SKOV-3 cells and a stronger anti-tumor efficacy with the tumor inhibition rate of 85.24%. The pharmacokinetics results of ES-SSL-OXA/PTX showed a prolonged half-life time and a slowed clearance rate. The preliminary safety study of acute toxicity and long-term toxicity demonstrated ES-SSL-OXA/PTX exhibited a reduced toxicity profile. Based on the above results, ES-SSL-OXA/PTX could be a promising novel formulation for the treatment of ovarian cancer in future clinic.
Collapse
Affiliation(s)
- Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhihui Ren
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongyu Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Zhe Lv
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yan Zhang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Rui Liu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yujia Shen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Yucui Zheng
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Dongfanghui Miao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xin Guo
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Hongli Chen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Shanshan Wang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
30
|
Current understanding of passive and active targeting nanomedicines to enhance tumor accumulation. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
31
|
Du M, Geng T, Yu R, Song G, Cheng H, Cao Y, He W, Haleem A, Li Q, Hu R, Chen S. Smart anti-vascular nanoagent induces positive feedback loop for self-augmented tumor accumulation. J Control Release 2023; 356:595-609. [PMID: 36924896 DOI: 10.1016/j.jconrel.2023.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023]
Abstract
How to achieve efficient drug accumulation in the tumor with low vascular density is a great challenge but the key to push the limit of anti-vascular therapeutic efficacy. Herein, we report a charge-reversible nanoparticles of gambogenic acid (CRNP-GNA) that would induce the positive feedback loop between increased tumor vascular permeability and improved drug accumulation. This positive feedback loop would remarkably improve tumor vascular permeability for efficient drug accumulation through few residue vessels. As compared to its charge-irreversible analogue in the latter injections, the accumulation in tumor and vascular permeability and retention indexes (VPRI) in CRNP-GNA group respectively boosted from nearly equal to 8.32 and 60 times, while its tumorous microvessel density decreased from nearly equal to only 7%. The self-augmented accumulation consequently amplified the antitumor efficacy via multiple pathways of anti-angiogenesis, vascular disruption and pro-apoptosis, where 5 out of 6 tumors in animal models were completely cured by CRNP-GNA. This work confirms that the underlying positive feedback loop for anti-vascular therapy could be induced by charge-reversible drug delivery nanosystem to achieve efficient and self-augmented drug accumulation even in the tumor with few vessels. It provides a novel strategy to conquer the dilemma between anti-vascular efficacy and drug accumulation.
Collapse
Affiliation(s)
- Mengting Du
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Tingting Geng
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Rongrong Yu
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Gang Song
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Hui Cheng
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Yu Cao
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Weidong He
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Abdul Haleem
- School of Chemistry and Chemical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Qinglin Li
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China
| | - Rongfeng Hu
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.
| | - Shengqi Chen
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Key Laboratory of Xin'an Medicine, The Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.
| |
Collapse
|
32
|
Subhan MA, Parveen F, Filipczak N, Yalamarty SSK, Torchilin VP. Approaches to Improve EPR-Based Drug Delivery for Cancer Therapy and Diagnosis. J Pers Med 2023; 13:jpm13030389. [PMID: 36983571 PMCID: PMC10051487 DOI: 10.3390/jpm13030389] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The innovative development of nanomedicine has promised effective treatment options compared to the standard therapeutics for cancer therapy. However, the efficiency of EPR-targeted nanodrugs is not always pleasing as it is strongly prejudiced by the heterogeneity of the enhanced permeability and retention effect (EPR). Targeting the dynamics of the EPR effect and improvement of the therapeutic effects of nanotherapeutics by using EPR enhancers is a vital approach to developing cancer therapy. Inadequate data on the efficacy of EPR in humans hampers the clinical translation of cancer drugs. Molecular targeting, physical amendment, or physiological renovation of the tumor microenvironment (TME) are crucial approaches for improving the EPR effect. Advanced imaging technologies for the visualization of EPR-induced nanomedicine distribution in tumors, and the use of better animal models, are necessary to enhance the EPR effect. This review discusses strategies to enhance EPR effect-based drug delivery approaches for cancer therapy and imaging technologies for the diagnosis of EPR effects. The effort of studying the EPR effect is beneficial, as some of the advanced nanomedicine-based EPR-enhancing approaches are currently undergoing clinical trials, which may be helpful to improve EPR-induced drug delivery and translation to clinics.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Correspondence: (M.A.S.); (V.P.T.)
| | - Farzana Parveen
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital Jhang 35200, Primary and Secondary Healthcare Department, Government of Punjab, Lahore, Punjab 54000, Pakistan
| | - Nina Filipczak
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Vladimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Correspondence: (M.A.S.); (V.P.T.)
| |
Collapse
|
33
|
Zhang D, He J, Zhou M. Radiation-assisted strategies provide new perspectives to improve the nanoparticle delivery to tumor. Adv Drug Deliv Rev 2023; 193:114642. [PMID: 36529190 DOI: 10.1016/j.addr.2022.114642] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/07/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022]
Abstract
Nanoparticles (NPs), with advantages in tumor targeting, have been extensively developed for anticancer treatment. However, the delivery efficacy of NPs tends to be heterogeneous in clinical research. Surprisingly, a traditional cancer treatment, radiotherapy (radiation), has been observed with the potential to improve the delivery of NPs by influencing the features of the tumor microenvironment, which provides new perspectives to overcome the barriers in the NPs delivery. Since the effect of radiation can also be enhanced by versatile NPs, these findings of radiation-assisted NPs delivery suggest innovative strategies combining radiotherapy with nanotherapeutics. This review summarizes the research on the delivery and therapeutic efficacy of NPs that are improved by radiation, focusing on relative mechanisms and existing challenges and opportunities.
Collapse
Affiliation(s)
- Dongxiao Zhang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Jian He
- The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining 314400, China; The Institute of Translational Medicine, Zhejiang University, Hangzhou 310029, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou 310053, China.
| |
Collapse
|
34
|
Bioactive lipid-nanoparticles with inherent self-therapeutic and anti-angiogenic properties for cancer therapy. Acta Biomater 2023; 157:500-510. [PMID: 36535568 DOI: 10.1016/j.actbio.2022.12.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/22/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Angiogenesis inhibition has become a promising therapeutical strategy for cancer treatment. Current clinical anti-angiogenesis treatment includes antibodies against vascular endothelial growth factor (VEGF) or VEGF receptor, fusion proteins with high affinity to VEGF receptor, and tyrosine kinase inhibitors of VEGF receptor. However, current treatments are prone to systemic toxicity or acquiring drug resistance. A natural bioactive lipid 1,2-dipalmitoyl-sn‑glycero-3-phosphate (dipalmitoyl phosphatidic acid, DPPA) was reported to exhibit anti-angiogenic and anti-tumoral activity. However, the hydrophobic property of DPPA largely restricted its clinical use, while systemic infusion of free DPPA could result in undesirable side effects. Herein, we successfully developed DPPA-based lipid-nanoparticles (DPPA-LNPs) which turns the "therapeutic payload into nanocarrier". This strategy could improve on DPPA's hydrophiliciy, thereby facilitating its systemic administration. . DPPA-LNPs not only retained the therapeutic anti-angiogenic and anti-tumoral bioactivity of parental DPPA, but also greatly improved its tumor targeting ability via enhanced permeability and retention (EPR) effect. This strategy not only eliminates the limitation of drug encapsulation rate, toxicity of the delivery vehicle; but also enhances DPPA bioacvtity in vitro and in vivo. Systemic administration of DPPA-LNPs significantly suppressed the blood vessel formation and tumor growth of triple negative breast cancer and liver cancer growth on both xenograft tumor models. STATEMENT OF SIGNIFICANCE: This is the first-in-kind self-therapeutic inherent lipid to be made into a nanocarrier, with inherent anti-angiogenic and anti-tumor properties. DPPA nanocarrier is fully natural, fully compatible with minimal systemic toxicity. DPPA nanocarrier can accumulate at high concentration at tumor via EPR effect, exerting both anti-angiogenic and anti-tumor effects in vivo. DPPA nanocarrier could be used to encapsulate biologics or small molecules for synergistic anti-cancer therapy.
Collapse
|
35
|
Tanaka HY, Nakazawa T, Enomoto A, Masamune A, Kano MR. Therapeutic Strategies to Overcome Fibrotic Barriers to Nanomedicine in the Pancreatic Tumor Microenvironment. Cancers (Basel) 2023; 15:cancers15030724. [PMID: 36765684 PMCID: PMC9913712 DOI: 10.3390/cancers15030724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Pancreatic cancer is notorious for its dismal prognosis. The enhanced permeability and retention (EPR) effect theory posits that nanomedicines (therapeutics in the size range of approximately 10-200 nm) selectively accumulate in tumors. Nanomedicine has thus been suggested to be the "magic bullet"-both effective and safe-to treat pancreatic cancer. However, the densely fibrotic tumor microenvironment of pancreatic cancer impedes nanomedicine delivery. The EPR effect is thus insufficient to achieve a significant therapeutic effect. Intratumoral fibrosis is chiefly driven by aberrantly activated fibroblasts and the extracellular matrix (ECM) components secreted. Fibroblast and ECM abnormalities offer various potential targets for therapeutic intervention. In this review, we detail the diverse strategies being tested to overcome the fibrotic barriers to nanomedicine in pancreatic cancer. Strategies that target the fibrotic tissue/process are discussed first, which are followed by strategies to optimize nanomedicine design. We provide an overview of how a deeper understanding, increasingly at single-cell resolution, of fibroblast biology is revealing the complex role of the fibrotic stroma in pancreatic cancer pathogenesis and consider the therapeutic implications. Finally, we discuss critical gaps in our understanding and how we might better formulate strategies to successfully overcome the fibrotic barriers in pancreatic cancer.
Collapse
Affiliation(s)
- Hiroyoshi Y. Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Takuya Nakazawa
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
| | - Atsushi Enomoto
- Department of Pathology, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya-shi 466-8550, Aichi, Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai-shi 980-8574, Miyagi, Japan
| | - Mitsunobu R. Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama-shi 700-8530, Okayama, Japan
- Correspondence:
| |
Collapse
|
36
|
Cheng X, Xie Q, Sun Y. Advances in nanomaterial-based targeted drug delivery systems. Front Bioeng Biotechnol 2023; 11:1177151. [PMID: 37122851 PMCID: PMC10133513 DOI: 10.3389/fbioe.2023.1177151] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Nanomaterial-based drug delivery systems (NBDDS) are widely used to improve the safety and therapeutic efficacy of encapsulated drugs due to their unique physicochemical and biological properties. By combining therapeutic drugs with nanoparticles using rational targeting pathways, nano-targeted delivery systems were created to overcome the main drawbacks of conventional drug treatment, including insufficient stability and solubility, lack of transmembrane transport, short circulation time, and undesirable toxic effects. Herein, we reviewed the recent developments in different targeting design strategies and therapeutic approaches employing various nanomaterial-based systems. We also discussed the challenges and perspectives of smart systems in precisely targeting different intravascular and extravascular diseases.
Collapse
|
37
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
38
|
Xu Y, Nie Z, Ni N, Zhang X, Yuan J, Gao Y, Gong Y, Liu S, Wu M, Sun X. Shield-activated two-way imaging nanomaterials for enhanced cancer theranostics. Biomater Sci 2022; 10:6893-6910. [PMID: 36317535 DOI: 10.1039/d2bm01317g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Smart nanomaterials with stimuli-responsive imaging enhancement have been widely developed to meet the requirements of accurate cancer diagnosis. However, these imaging nanoenhancers tend to be always on during circulation, which significantly increases the background signal when assessing the imaging performance. To improve unfavorable signal-to-noise ratios, an effective way is to shield the noise signal of these nanoprobes in non-targeted areas. Fortunately, there is a natural mutual shielding effect between some imaging nanomaterials, which provides the possibility of designing engineered nanomaterials with imaging quenching between two different components at the beginning. Once in the tumor microenvironment, the two components will present activated dual-mode imaging ability because of their separation, designated as two-way imaging tuning. This review highlights the design and mechanism of a series of engineered nanomaterials with two-way imaging tuning and their latest applications in the fields of cancer magnetic resonance imaging, fluorescence imaging, and their combination. The challenges and future directions for the improvement of these engineered nanomaterials towards clinical transformation are also discussed. This review aims to introduce the special constraint relationships of imaging components and provide scientists with simpler and more efficient nanoplatform construction ideas, promoting the development of engineered nanomaterials with two-way imaging tuning in cancer theranostics.
Collapse
Affiliation(s)
- Yang Xu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Zhaokun Nie
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Nengyi Ni
- Department of Chemical and Biomolecular Engineering, National University of Singapore, Singapore 117585, Singapore
| | - Xinyu Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Jia Yuan
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Yuan Gao
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Yufang Gong
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Shuangqing Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| | - Min Wu
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250000, China.
| |
Collapse
|
39
|
Liu HJ, Xu P. Strategies to overcome/penetrate the BBB for systemic nanoparticle delivery to the brain/brain tumor. Adv Drug Deliv Rev 2022; 191:114619. [PMID: 36372301 PMCID: PMC9724744 DOI: 10.1016/j.addr.2022.114619] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/23/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
Despite its prevalence in the management of peripheral tumors, compared to surgery and radiation therapy, chemotherapy is still a suboptimal intervention in fighting against brain cancer and cancer brain metastases. This discrepancy is mainly derived from the complicatedly physiological characteristic of intracranial tumors, including the presence of blood-brain barrier (BBB) and limited enhanced permeability and retention (EPR) effect attributed to blood-brain tumor barrier (BBTB), which largely lead to insufficient therapeutics penetrating to tumor lesions to produce pharmacological effects. Therefore, dependable methodologies that can boost the efficacy of chemotherapy for brain tumors are urgently needed. Recently, nanomedicines have shown great therapeutic potential in brain tumors by employing various transcellular strategies, paracellular strategies, and their hybrids, such as adsorptive-mediated transcytosis, receptor-mediated transcytosis, BBB disruption technology, and so on. It is compulsory to comprehensively summarize these practices to shed light on future directions in developing therapeutic regimens for brain tumors. In this review, the biological and pathological characteristics of brain tumors, including BBB and BBTB, are illustrated. After that, the emerging delivery strategies for brain tumor management are summarized into different classifications and supported with detailed examples. Finally, the potential challenges and prospects for developing and clinical application of brain tumor-oriented nanomedicine are discussed.
Collapse
Affiliation(s)
- Hai-Jun Liu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA
| | - Peisheng Xu
- Department of Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC 29208, USA.
| |
Collapse
|
40
|
Peng T, Xu T, Liu X. Research progress of the engagement of inorganic nanomaterials in cancer immunotherapy. Drug Deliv 2022; 29:1914-1932. [PMID: 35748543 PMCID: PMC9246104 DOI: 10.1080/10717544.2022.2086940] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer has attracted widespread attention from scientists for its high morbidity and mortality, posing great threats to people’s health. Cancer immunotherapy with high specificity, low toxicity as well as triggering systemic anti-tumor response has gradually become common in clinical cancer treatment. However, due to the insufficient immunogenicity of tumor antigens peptides, weak ability to precisely target tumor sites, and the formation of tumor immunosuppressive microenvironment, the efficacy of immunotherapy is often limited. In recent years, the emergence of inorganic nanomaterials makes it possible for overcoming the limitations mentioned above. With self-adjuvant properties, high targeting ability, and good biocompatibility, the inorganic nanomaterials have been integrated with cancer immunotherapy and significantly improved the therapeutic effects.
Collapse
Affiliation(s)
- Tingwei Peng
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Pudong New Area, China
| | - Tianzhao Xu
- Shanghai Qiansu Biological Technology Co., Ltd, Pudong New Area, China.,Department of Clinical Laboratory, Gongli Hospital, School of Medicine, Shanghai University, Shanghai, China
| | - Xinghui Liu
- Department of Clinical Laboratory, Gongli Hospital, School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
41
|
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: 6] [Impact Index Per Article: 3.0] [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
|
42
|
Youden B, Jiang R, Carrier AJ, Servos MR, Zhang X. A Nanomedicine Structure-Activity Framework for Research, Development, and Regulation of Future Cancer Therapies. ACS NANO 2022; 16:17497-17551. [PMID: 36322785 DOI: 10.1021/acsnano.2c06337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Despite their clinical success in drug delivery applications, the potential of theranostic nanomedicines is hampered by mechanistic uncertainty and a lack of science-informed regulatory guidance. Both the therapeutic efficacy and the toxicity of nanoformulations are tightly controlled by the complex interplay of the nanoparticle's physicochemical properties and the individual patient/tumor biology; however, it can be difficult to correlate such information with observed outcomes. Additionally, as nanomedicine research attempts to gradually move away from large-scale animal testing, the need for computer-assisted solutions for evaluation will increase. Such models will depend on a clear understanding of structure-activity relationships. This review provides a comprehensive overview of the field of cancer nanomedicine and provides a knowledge framework and foundational interaction maps that can facilitate future research, assessments, and regulation. By forming three complementary maps profiling nanobio interactions and pathways at different levels of biological complexity, a clear picture of a nanoparticle's journey through the body and the therapeutic and adverse consequences of each potential interaction are presented.
Collapse
Affiliation(s)
- Brian Youden
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Runqing Jiang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Medical Physics, Grand River Regional Cancer Centre, Kitchener, Ontario N2G 1G3, Canada
| | - Andrew J Carrier
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| | - Mark R Servos
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
| | - Xu Zhang
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada
- Department of Chemistry, Cape Breton University, 1250 Grand Lake Road, Sydney, Nova Scotia B1P 6L2, Canada
| |
Collapse
|
43
|
Jiang Z, Zhang W, Zhang J, Liu T, Xing J, Zhang H, Tang D. Nanomaterial-Based Drug Delivery Systems: A New Weapon for Cancer Immunotherapy. Int J Nanomedicine 2022; 17:4677-4696. [PMID: 36211025 PMCID: PMC9541303 DOI: 10.2147/ijn.s376216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer immunotherapy, a major breakthrough in cancer treatment, has been successfully applied to treat a number of tumors. However, given the presence of factors in the tumor microenvironment (TME) that impede immunotherapy, only a small proportion of patients achieve a good clinical response. With the ability to increase permeability and cross biological barriers, nanomaterials have been successfully applied to deliver immunotherapeutic agents, thus realizing the anti-cancer therapeutic potential of therapeutic agents. This has driven a wave of research into systems for the delivery of immunotherapeutic agents, which has resulted in widespread interest in nanomaterial-based drug delivery systems. Nanomaterial-based drug delivery systems are able to overcome the challenges from TME and thus achieve good results in cancer immunotherapy. If it can make a breakthrough in improving biocompatibility and reducing cytotoxicity, it will be more widely used in clinical practice. Different types of nanomaterials may also have some subtle differences in enhancing cancer immunotherapy. Moreover, delivery systems made of nanomaterials loaded with drugs, such as cytotoxic drugs, cytokines, and adjuvants, could be used for cancer immunotherapy because they avoid the toxicity and side effects associated with these drugs, thereby enabling their reuse. Therefore, further insights into nanomaterial-based drug delivery systems will provide more effective treatment options for cancer patients.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China,Correspondence: Dong Tang, Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, 225000, People’s Republic of China, Email
| |
Collapse
|
44
|
Abstract
The field of cancer nanomedicine seeks to overcome the inherent shortcomings of conventional cancer diagnostics and therapies. Yet despite the surge of interest in and attractive attributes of nanotechnologies, challenges remain in their clinical translation, prompting some to argue that they have not yet reached their true potential. In this Viewpoint article, we asked four experts for their opinions on how we can fulfil the great promise of nanomedicine for the detection, diagnosis and treatment of patients with cancer.
Collapse
Affiliation(s)
- Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Department of Chemical and Biomolecular Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), RWTH Aachen University Clinic, Aachen, Germany.
| |
Collapse
|
45
|
Smart pH-responsive polyhydralazine/bortezomib nanoparticles for remodeling tumor microenvironment and enhancing chemotherapy. Biomaterials 2022; 288:121737. [PMID: 36031455 DOI: 10.1016/j.biomaterials.2022.121737] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/01/2022] [Accepted: 08/06/2022] [Indexed: 01/01/2023]
Abstract
The clinical translation of nanomedicines has been impeded by the unfavorable tumor microenvironment (TME), particularly the tortuous vasculature networks, which significantly influence the transport and distribution of nanomedicines into tumors. In this work, a smart pH-responsive bortezomib (BTZ)-loaded polyhydralazine nanoparticle (PHDZ/BTZ) is presented, which has a great capacity to augment the accumulation of BTZ in tumors by dilating tumor blood vessels via specific release of vasodilator hydralazine (HDZ). The Lewis acid-base coordination effect between the boronic bond of BTZ and amino of HDZ empowered PHDZ/BTZ nanoparticles with great stability and high drug loading contents. Once triggered by the acidic tumor environment, HDZ could be released quickly to remodel TME through tumor vessel dilation, hypoxia attenuation, and lead to an increased intratumoral BTZ accumulation. Additionally, our investigation revealed that this pH-responsive nanoparticle dramatically suppressed tumor growth, inhibited the occurrence of lung metastasis with fewer side effects and induced immunogenic cell death (ICD), thereby eliciting immune activation including massive cytotoxic T lymphocytes (CTLs) infiltration in tumors and efficient serum proinflammatory cytokine secretion compared with free BTZ treatment. Thus, with efficient drug loading capacity and potent immune activation, PHDZ nanoparticles exhibit great potential in the delivery of boronic acid-containing drugs aimed at a wide range of diseases.
Collapse
|
46
|
Ren M, Zheng X, Gao H, Jiang A, Yao Y, He W. Nanomedicines Targeting Metabolism in the Tumor Microenvironment. Front Bioeng Biotechnol 2022; 10:943906. [PMID: 35992338 PMCID: PMC9388847 DOI: 10.3389/fbioe.2022.943906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/01/2022] [Indexed: 12/02/2022] Open
Abstract
Cancer cells reprogram their metabolism to meet their growing demand for bioenergy and biosynthesis. The metabolic profile of cancer cells usually includes dysregulation of main nutritional metabolic pathways and the production of metabolites, which leads to a tumor microenvironment (TME) having the characteristics of acidity, hypoxic, and/or nutrient depletion. Therapies targeting metabolism have become an active and revolutionary research topic for anti-cancer drug development. The differential metabolic vulnerabilities between tumor cells and other cells within TME provide nanotechnology a therapeutic window of anti-cancer. In this review, we present the metabolic characteristics of intrinsic cancer cells and TME and summarize representative strategies of nanoparticles in metabolism-regulating anti-cancer therapy. Then, we put forward the challenges and opportunities of using nanoparticles in this emerging field.
Collapse
Affiliation(s)
- Mengdi Ren
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqiang Zheng
- Institute for Stem Cell and Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Huan Gao
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Aimin Jiang
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yu Yao
- Department of Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yu Yao, ; Wangxiao He,
| | - Wangxiao He
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Yu Yao, ; Wangxiao He,
| |
Collapse
|
47
|
Zi Y, Yang K, He J, Wu Z, Liu J, Zhang W. Strategies to enhance drug delivery to solid tumors by harnessing the EPR effects and alternative targeting mechanisms. Adv Drug Deliv Rev 2022; 188:114449. [PMID: 35835353 DOI: 10.1016/j.addr.2022.114449] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/03/2022] [Accepted: 07/06/2022] [Indexed: 12/20/2022]
Abstract
The Enhanced Permeability and Retention (EPR) effect has been recognized as the central paradigm in tumor-targeted delivery in the last decades. In the wake of this concept, nanotechnologies have reached phenomenal levels in research. However, clinical tumors display a poor manifestation of EPR effect. Factors including tumor heterogeneity, complicating tumor microenvironment, and discrepancies between laboratory models and human tumors largely contribute to poor efficiency in tumor-targeted delivery and therapeutic failure in clinical translation. In this article, approaches for evaluation of EPR effect in human tumor were overviewed as guidance to employ EPR effect for cancer treatment. Strategies to augment EPR-mediated tumoral delivery are discussed in different dimensions including enhancement of vascular permeability, depletion of tumor extracellular matrix and optimization of nanoparticle design. Besides, the recent development in alternative tumor-targeted delivery mechanisms are highlighted including transendothelial pathway, endogenous cell carriers and non-immunogenic bacteria-mediated delivery. In addition, the emerging preclinical models better reflect human tumors are introduced. Finally, more rational applications of EPR effect in other disease and field are proposed. This article elaborates on fundamental reasons for the gaps between theoretical expectation and clinical outcomes, attempting to provide some perspective directions for future development of cancer nanomedicines in this still evolving landscape.
Collapse
Affiliation(s)
- Yixuan Zi
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Kaiyun Yang
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | - Jianhua He
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China
| | - Zimei Wu
- School of Pharmacy, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | - Jianping Liu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
48
|
Approaches to Improve Macromolecule and Nanoparticle Accumulation in the Tumor Microenvironment by the Enhanced Permeability and Retention Effect. Polymers (Basel) 2022; 14:polym14132601. [PMID: 35808648 PMCID: PMC9268820 DOI: 10.3390/polym14132601] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/17/2022] Open
Abstract
Passive targeting is the foremost mechanism by which nanocarriers and drug-bearing macromolecules deliver their payload selectively to solid tumors. An important driver of passive targeting is the enhanced permeability and retention (EPR) effect, which is the cornerstone of most carrier-based tumor-targeted drug delivery efforts. Despite the huge number of publications showcasing successes in preclinical animal models, translation to the clinic has been poor, with only a few nano-based drugs currently being used for the treatment of cancers. Several barriers and factors have been adduced for the low delivery efficiency to solid tumors and poor clinical translation, including the characteristics of the nanocarriers and macromolecules, vascular and physiological barriers, the heterogeneity of tumor blood supply which affects the homogenous distribution of nanocarriers within tumors, and the transport and penetration depth of macromolecules and nanoparticles in the tumor matrix. To address the challenges associated with poor tumor targeting and therapeutic efficacy in humans, the identified barriers that affect the efficiency of the enhanced permeability and retention (EPR) effect for macromolecular therapeutics and nanoparticle delivery systems need to be overcome. In this review, approaches to facilitate improved EPR delivery outcomes and the clinical translation of novel macromolecular therapeutics and nanoparticle drug delivery systems are discussed.
Collapse
|
49
|
Zhou Y, Xu L, Wang Z, Liu H, Zhang X, Shu C, Zhang M, Wang T, Xu X, Pu X, He J, Wang P, Qiu Y, Xu G, Zou X, Zhu Y, Wang L. Sequentially targeting and intervening mutual Polo-like Kinase 1 on CAFs and tumor cells by dual targeting nano-platform for cholangiocarcinoma treatment. Theranostics 2022; 12:3911-3927. [PMID: 35664077 PMCID: PMC9131280 DOI: 10.7150/thno.70557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/26/2022] [Indexed: 11/18/2022] Open
Abstract
Rationale: Synergistic treatment strategies for two or more drugs have gradually developed as the main options in clinics for cholangiocarcinoma (CCA) owing to the complicated crosstalk between the tumor and stroma. However, the different synergetic mechanisms pose great challenges to the dosages and order of administration of drugs. Thus, a strategy for exploring and intervening in mutual targets derived from stromal cells and cholangiocarcinoma cells was proposed. Methods: Genes with overexpression patterns in tumors and displaying a significant association with overall survival were identified from RNA-seq data of human CCA patients and CCA mouse models. Western blotting, qRT-PCR, immunofluorescence (IF), colony formation and flow cytometry assays were conducted to determine the biological roles of the key oncogene in cholangiocarcinoma and stromal cells respectively. Additionally, a dual-targeting drug delivery system (AA-HA-ODA) for cancer-associated fibroblasts (CAFs) and tumor cells was constructed to verify the effectiveness of intervening the screened genes in vivo. Results: Polo-like kinase 1 (PLK1) was verified to play vital role in the malignant proliferation of CCA by regulating the cell cycle pathway. PLK1 also decreased stromal production by regulating the CAF phenotype. In addition, a PLK1 inhibitor (Ro3280) loaded dual-targeting drug delivery system (AA-HA-ODA) was prepared and exhibited high affinity for CAFs and cholangiocarcinoma cells. The in vivo distribution pattern and antitumor efficacy of AA-HA-ODA/Ro also verify the effectiveness of inhibiting PLK1 in CCA in vivo. Conclusion: In summary, PLK1 is a mutual target derived from tumor cells and stroma due to its crucial role in the proliferation of tumor cells and stroma regulation in CAFs, which might provide enlightenment for multitarget treatment strategies and guidance for clinical cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Zhangding Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Hongwen Liu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xiang Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Chuanjun Shu
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 211166, Jiangsu Province, China
| | - Meng Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Ting Wang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Xinyun Xu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Pin Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Yudong Qiu
- Department of Hepatopancreatobiliary Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
| | - Guifang Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xiaoping Zou
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Yun Zhu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
- Department of Pharmacy, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, Jiangsu Province, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu Province, China
- Department of Gastroenterology, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| |
Collapse
|
50
|
Hashida M. Advocation and advancements of EPR effect theory in drug delivery science: A commentary. J Control Release 2022; 346:355-357. [PMID: 35483640 DOI: 10.1016/j.jconrel.2022.04.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 10/18/2022]
Abstract
To honor the contributions of Professor Hiroshi Maeda to the progress of targeted drug delivery research, a brief review of enhanced permeability and retention (EPR) effect theory proposed by him as the physiology-based principal mechanism of intra-tumoral accumulation of large molecules and small particles is presented. Under historical and practical backgrounds in developments of various drug delivery systems including macromolecular conjugates, the concept of EPR effect was advocated in mid1980s and has cultivated new cancer chemotherapeutic modalities until recently. Namely, nanoplatforms such as polymer conjugates, liposomes, polymeric micelles, and nanoparticles have been studied as a promising fusion area for nanotechnology and medicine. Modulation of EPR effect by chemical and/or mechanical approaches to achieve tumor vascular and tissue modification would further lead to sophistication of cancer chemotherapy employing nanomedicines.
Collapse
Affiliation(s)
- Mitsuru Hashida
- Institute for Integrated Cell-Materia Sciences and Graduate School of Pharmaceutical Sciences, Kyoto University, Yoshidaushinomiya-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| |
Collapse
|