1
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [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: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
2
|
Zhang B, Yan J, Jin Y, Yang Y, Zhao X. Curcumin-shellac nanoparticle-loaded GelMA/SilMA hydrogel for colorectal cancer therapy. Eur J Pharm Biopharm 2024; 202:114409. [PMID: 38996942 DOI: 10.1016/j.ejpb.2024.114409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
In this study, a novel approach was employed to develop a therapeutic system for colorectal cancer treatment. Specifically, a GelMA/SilMA hydrogel loaded with curcumin-shellac nanoparticles (Cur@Lac NPs) was created. A microfluidic swirl mixer was utilized to formulate stable Cur@Lac NPs, ensuring their consistent and effective encapsulation. The pH-specific release of curcumin from the NPs demonstrated their potential for colon cancer treatment. By carefully regulating the ratio of GelMA (gelatin methacrylate) and SilMA (silk fibroin methacrylate), a GelMA/SilMA dual network hydrogel was generated, offering controlled release and degradation capabilities. The incorporation of SilMA notably enhanced the mechanical properties of the dual network matrix, improving compression resistance and mitigating deformation. This mechanical improvement is crucial for maintaining the structural integrity of the hydrogel during in vivo applications. In comparison to the direct incubation of curcumin, the strategy of encapsulating curcumin into NPs and embedding them within the GelMA/SilMA hydrogel resulted in more controlled release mechanisms. This controlled release was achieved through the disintegration of the NPs and the swelling and degradation of the hydrogel matrix. The encapsulating strategy also demonstrated enhanced cellular uptake of curcumin, leveraging the advantages of both NPs and in-situ hydrogel injection. This combination ensures a more efficient and sustained delivery of the therapeutic agent directly to the tumor site. Overall, this approach holds significant promise as a smart drug delivery system, potentially improving the efficacy of colorectal cancer treatments by providing targeted, controlled, and sustained drug release with enhanced mechanical stability and biocompatibility.
Collapse
Affiliation(s)
- Bo Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Jiaxuan Yan
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yi Jin
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou 213000, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China
| | - Yushun Yang
- Jinhua Advanced Research Institute, Jinhua 321019, China
| | - Xiubo Zhao
- School of Pharmacy, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
3
|
Cuoghi S, Caraffi R, Anderlini A, Baraldi C, Enzo E, Vandelli MA, Tosi G, Ruozi B, Duskey JT, Ottonelli I. Challenges of enzyme therapy: Why two players are better than one. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1979. [PMID: 38955512 DOI: 10.1002/wnan.1979] [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: 01/02/2024] [Revised: 04/29/2024] [Accepted: 05/28/2024] [Indexed: 07/04/2024]
Abstract
Enzyme-based therapy has garnered significant attention for its current applications in various diseases. Despite the notable advantages associated with the use of enzymes as therapeutic agents, that could have high selectivity, affinity, and specificity for the target, their application faces challenges linked to physico-chemical and pharmacological properties. These limitations can be addressed through the encapsulation of enzymes in nanoplatforms as a comprehensive solution to mitigate their degradation, loss of activity, off-target accumulation, and immunogenicity, thus enhancing bioavailability, therapeutic efficacy, and circulation time, thereby reducing the number of administrations, and ameliorating patient compliance. The exploration of novel nanomedicine-based enzyme therapeutics for the treatment of challenging diseases stands as a paramount goal in the contemporary scientific landscape, but even then it is often not enough. Combining an enzyme with another therapeutic (e.g., a small molecule, another enzyme or protein, a monoclonal antibody, or a nucleic acid) within a single nanocarrier provides innovative multidrug-integrated therapy and ensures that both the actives arrive at the target site and exert their therapeutic effect, leading to synergistic action and superior therapeutic efficacy. Moreover, this strategic approach could be extended to gene therapy, a field that nowadays has gained increasing attention, as enzymes acting at genomic level and nucleic acids may be combined for synergistic therapy. This multicomponent therapeutic approach opens opportunities for promising future developments. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Sabrina Cuoghi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Riccardo Caraffi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Anderlini
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Baraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Enzo
- Centre for Regenerative Medicine "Stefano Ferrari", University of Modena and Reggio Emilia, Modena, Italy
| | - Maria Angela Vandelli
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Barbara Ruozi
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ilaria Ottonelli
- Nanotech Lab, Te.Far.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
4
|
Dosta P, Dion MZ, Prado M, Hurtado P, Riojas-Javelly CJ, Cryer AM, Soria Y, Andrews Interiano N, Muñoz-Taboada G, Artzi N. Matrix Metalloproteinase- and pH-Sensitive Nanoparticle System Enhances Drug Retention and Penetration in Glioblastoma. ACS NANO 2024; 18:14145-14160. [PMID: 38761153 DOI: 10.1021/acsnano.3c03409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Glioblastoma (GBM) is a primary malignant brain tumor with limited therapeutic options. One promising approach is local drug delivery, but the efficacy is hindered by limited diffusion and retention. To address this, we synthesized and developed a dual-sensitive nanoparticle (Dual-NP) system, formed between a dendrimer and dextran NPs, bound by a dual-sensitive [matrix metalloproteinase (MMP) and pH] linker designed to disassemble rapidly in the tumor microenvironment. The disassembly prompts the in situ formation of nanogels via a Schiff base reaction, prolonging Dual-NP retention and releasing small doxorubicin (Dox)-conjugated dendrimer NPs over time. The Dual-NPs were able to penetrate deep into 3D spheroid models and detected at the tumor site up to 6 days after a single intratumoral injection in an orthotopic mouse model of GBM. The prolonged presence of Dual-NPs in the tumor tissue resulted in a significant delay in tumor growth and an overall increase in survival compared to untreated or Dox-conjugated dendrimer NPs alone. This Dual-NP system has the potential to deliver a range of therapeutics for efficiently treating GBM and other solid tumors.
Collapse
Affiliation(s)
- Pere Dosta
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Michelle Z Dion
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- MIT-Harvard Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Michaela Prado
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Engineering and Sciences, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Pau Hurtado
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Cristobal J Riojas-Javelly
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Engineering and Sciences, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | - Alexander M Cryer
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yael Soria
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Nelly Andrews Interiano
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Department of Engineering and Sciences, Tecnológico de Monterrey, Monterrey 64849, Mexico
| | | | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
- BioDevek Inc., Allston, Massachusetts 02134, United States
| |
Collapse
|
5
|
Lai C, Lin S, Liu W, Jin Y. Research Progress of Chitosan-based Multifunctional Nanoparticles in Cancer Targeted Therapy. Curr Med Chem 2024; 31:3074-3092. [PMID: 37062062 DOI: 10.2174/0929867330666230416153352] [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: 10/20/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 04/17/2023]
Abstract
Conventional tumor therapeutic modalities, such as radiotherapy, chemotherapy, and surgery, involve low tumor inhibition efficiency, non-targeted drug delivery, and side effects. The development of novel and practical nano-drug delivery systems (DDSs) for targeted tumor therapy has become particularly important. Among various bioactive nanoparticles, chitosan is considered a suitable candidate for drug delivery due to its nontoxicity, good biocompatibility, and biodegradability. The amino and hydroxyl groups of chitosan endow it with the diverse function of chemical modification, thereby improving its physical and biological properties to meet the requirements of advanced biomedical applications. Therefore, it is necessary to review the property and applications of chitosan- based materials in biomedicine. In this review, the characteristics of chitosan related to its applications are first introduced, and then the preparation and modification of chitosan-based nanoparticles, including the function tailoring of chitosan-modified nanoparticles, are demonstrated and discussed. Finally, the opportunities and challenges of chitosan- based nanomaterials in this emerging field are proposed from the perspective of the rational and systematic design for the biomedicine field.
Collapse
Affiliation(s)
- Chunmei Lai
- College of Materials Science and Engineering, Fuzhou University, Fuzhou, 350108, China
- College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Simin Lin
- College of Materials Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Wei Liu
- Fujian College Association Instrumental Analysis Center of Fuzhou University, Fuzhou University, Fuzhou, 350108, China
| | - Yanqiao Jin
- College of Materials Science and Engineering, Fuzhou University, Fuzhou, 350108, China
| |
Collapse
|
6
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|
7
|
Indhu AR, Keerthana L, Dharmalingam G. Plasmonic nanotechnology for photothermal applications - an evaluation. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:380-419. [PMID: 37025366 PMCID: PMC10071519 DOI: 10.3762/bjnano.14.33] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
The application of plasmonic nanoparticles is motivated by the phenomenon of surface plasmon resonance. Owing to the tunability of optothermal properties and enhanced stability, these nanostructures show a wide range of applications in optical sensors, steam generation, water desalination, thermal energy storage, and biomedical applications such as photothermal (PT) therapy. The PT effect, that is, the conversion of absorbed light to heat by these particles, has led to thriving research regarding the utilization of plasmonic nanoparticles for a myriad of applications. The design of conventional nanomaterials for PT conversion has focussed predominantly on the manipulation of photon absorption through bandgap engineering, doping, incorporation, and modification of suitable matrix materials. Plasmonic nanomaterials offer an alternative and attractive approach in this regard, through the flexibility in the excitation of surface plasmons. Specific advantages are the considerable improved bandwidth of the absorption, a higher efficiency of photon absorption, facile tuning, as well as flexibility in the synthesis of plasmonic nanomaterials. This review of plasmonic PT (PPT) research begins with a theoretical discussion on the plasmonic properties of nanoparticles by means of the quasi-static approximation, Mie theory, Gans theory, generic simulations on common plasmonic material morphologies, and the evaluation processes of PT performance. Further, a variety of nanomaterials and material classes that have potential for PPT conversion are elucidated, such as plasmonic metals, bimetals, and metal-metal oxide nanocomposites. A detailed investigation of the essential, but often ignored, concept of thermal, chemical, and aggregation stability of nanoparticles is another part of this review. The challenges that remain, as well as prospective directions and chemistries, regarding nanomaterials for PT conversion are pondered on in the final section of the article, taking into account the specific requirements from different applications.
Collapse
Affiliation(s)
- A R Indhu
- Plasmonic Nanomaterials Laboratory, PSG Institute of Advanced Studies, Coimbatore-641004, India
| | - L Keerthana
- Plasmonic Nanomaterials Laboratory, PSG Institute of Advanced Studies, Coimbatore-641004, India
| | | |
Collapse
|
8
|
Promises and challenges for targeting the immunological players in the tumor micro-environment – Critical determinants for NP-based therapy. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
9
|
Zhang W, Zheng X. Patient-derived xenografts or organoids in the discovery of traditional and self-assembled drug for tumor immunotherapy. Front Oncol 2023; 13:1122322. [PMID: 37081982 PMCID: PMC10110942 DOI: 10.3389/fonc.2023.1122322] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/24/2023] [Indexed: 04/22/2023] Open
Abstract
In addition to the rapid development of immune checkpoint inhibitors, there has also been a surge in the development of self-assembly immunotherapy drugs. Based on the immune target, traditional tumor immunotherapy drugs are classified into five categories, namely immune checkpoint inhibitors, direct immune modulators, adoptive cell therapy, oncolytic viruses, and cancer vaccines. Additionally, the emergence of self-assembled drugs with improved precision and environmental sensitivity offers a promising innovation approach to tumor immunotherapy. Despite rapid advances in tumor immunotherapy drug development, all candidate drugs require preclinical evaluation for safety and efficacy, and conventional evaluations are primarily conducted using two-dimensional cell lines and animal models, an approach that may be unsuitable for immunotherapy drugs. The patient-derived xenograft and organoids models, however, maintain the heterogeneity and immunity of the pathological tumor heterogeneity.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Talent Highland, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqiang Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Xiaoqiang Zheng,
| |
Collapse
|
10
|
Xie L, Li J, Wang L, Dai Y. Engineering metal‐phenolic networks for enhancing cancer therapy by tumor microenvironment modulation. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 15:e1864. [PMID: 36333962 DOI: 10.1002/wnan.1864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/06/2022]
Abstract
The complicated tumor microenvironment (TME) is featured by low pH values, high redox status, and hypoxia, which greatly supports the genesis, development, and metastasis of tumors, leading to drug resistance and clinical failure. Moreover, a lot of immunosuppressive cells infiltrate in such TME, resulting in depressing immunotherapy. Therefore, the development of TME-responsive nanoplatforms has shown great significance in enhancing cancer therapeutics. Metal-phenolic networks (MPNs)-based nanosystems, which self-assemble via coordination of phenolic materials and metal ions, have emerged as excellent TME theranostic nanoplatforms. MPNs have unique properties including fast preparation, tunable morphologies, pH response, and biocompatibility. Besides, functionalization and surface modification can endow MPNs with specific functions for application requirements. Here, the representative engineering strategies of various polyphenols are first introduced, followed by the introduction of the engineering mechanisms of polyphenolic nanosystems, fabrication, and distinct properties of MPNs. Then, their advances in TME modulation are highlighted, such as antiangiogenesis, hypoxia relief, combination therapy sensitization, and immunosuppressive TME reversion. Finally, we will discuss the challenges and future perspectives of MPNs-based nanosystems for enhancing cancer therapy. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Lisi Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat‐Sen Memorial Hospital, Sun Yat‐Sen University Guangzhou China
| | - Jie Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM) Nanjing Tech University (NanjingTech) Nanjing China
| | - Leyu Wang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Biomaterials Research Center, School of Biomedical Engineering Southern Medical University Guangzhou Guangdong China
| | - Yunlu Dai
- Cancer Center and Institute of Translational Medicine, Faculty of Health Sciences University of Macau Macau China
- MOE Frontiers Science Center for Precision Oncology University of Macau Macau China
| |
Collapse
|
11
|
Pęczek P, Gajda M, Rutkowski K, Fudalej M, Deptała A, Badowska-Kozakiewicz AM. Cancer-associated inflammation: pathophysiology and clinical significance. J Cancer Res Clin Oncol 2022; 149:2657-2672. [PMID: 36260158 PMCID: PMC9579684 DOI: 10.1007/s00432-022-04399-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022]
Abstract
Purpose Cancer cells, despite stemming from the own cells of their host, usually elicit an immune response. This response usually enables elimination of cancer at its earliest stages. However, some tumors develop mechanisms of escaping immune destruction and even profiting from tumor-derived inflammation. Methods We summarized the roles of different immune cell populations in various processes associated with cancer progression and possible methods of reshaping tumor-associated inflammation to increase the efficacy of cancer therapy. Results Changes in various signaling pathways result in attraction of immunosuppressive, pro-tumorigenic cells, such as myeloid-derived suppressor cells, tumor-associated macrophages, and neutrophils, while at the same time suppressing the activity of lymphocytes, which have the potential of destroying cancer cells. These changes promote tumor progression by increasing angiogenesis and growth, accelerating metastasis, and impairing drug delivery to the tumor site. Conclusion Due to its multi-faceted role in cancer, tumor-associated inflammation can serve as a valuable therapy target. By increasing it, whether through decreasing overall immunosuppression with immune checkpoint inhibitor therapy or through more specific methods, such as cancer vaccines, oncolytic viruses, or chimeric antigen receptor T cells, cancer-derived immunosuppression can be overcome, resulting in immune system destroying cancer cells. Even changes occurring in the microbiota can influence the shape of antitumor response, which could provide new attractive diagnostic or therapeutic methods. Interestingly, also decreasing the distorted tumor-associated inflammation with non-steroidal anti-inflammatory drugs can lead to positive outcomes.
Collapse
Affiliation(s)
- Piotr Pęczek
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Gajda
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Kacper Rutkowski
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Marta Fudalej
- Department of Cancer Prevention, Medical University of Warsaw, Erazma Ciołka 27, Warsaw, Poland.,Department of Oncology and Haematology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | - Andrzej Deptała
- Department of Cancer Prevention, Medical University of Warsaw, Erazma Ciołka 27, Warsaw, Poland.,Department of Oncology and Haematology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | | |
Collapse
|
12
|
Wu C, Shen Z, Lu Y, Sun F, Shi H. p53 Promotes Ferroptosis in Macrophages Treated with Fe 3O 4 Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2022; 14:42791-42803. [PMID: 36112832 DOI: 10.1021/acsami.2c00707] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Fe3O4 nanoparticles are the most widely used magnetic nanoparticles in the biomedicine field. The biodistribution of most nanoparticles in vivo is determined by the capture of macrophages; however, the effects of nanoparticles on macrophages remain poorly understood. Here, we demonstrated that Fe3O4 nanoparticles could reduce macrophage viability after 48 h of treatment and induce a shift in macrophage polarization toward the M1 phenotype; RNA sequencing revealed the activation of the ferroptosis pathway and p53 upregulation compared to the control group. The expression in p53, xCT, glutathione peroxidase 4 (GPX4), and transferrin receptor (TFR) in macrophages was similar to that in erastin-induced ferroptosis in macrophages, and the ultrastructural morphology of mitochondria was consistent with that of erastin-treated cells. We used DCFH-DA to estimate the intracellular reactive oxygen species content in Fe3O4 nanoparticles treated with Ana-1 and JC-1 fluorescent probes to detect the mitochondrial membrane potential change; both showed to be time-dependent. Fer-1 inhibited the reduction of the glutathione/oxidized glutathione (GSH/GSSG) ratio and inhibited intracellular oxidative stress states; therefore, Fe3O4 nanoparticles induced ferroptosis in macrophages. Finally, we used pifithrin-α hydrobromide (PFT) as a p53 inhibitor to verify whether the high expression of p53 is involved in mediating this process. After PFT treatment, the live/dead cell rate, TFR, p53 expression, and GPX4 consumption were inhibited and mitigated the GSH/GSSG ratio reduction as well. This indicates that p53 may contribute to Fe3O4 nanoparticle-induced ferroptosis of macrophages. We provide a theoretical basis for the molecular mechanisms of ferroptosis in macrophages and the biotoxicity in vivo induced by Fe3O4 nanoparticles.
Collapse
Affiliation(s)
- Cong Wu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Zhiming Shen
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Yi Lu
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Fei Sun
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| | - Hongcan Shi
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225000, China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
13
|
Chen W, Wang J, Yang H, Sun Y, Chen B, Liu Y, Han Y, Shan M, Zhan J. Interleukin 22 and its association with neurodegenerative disease activity. Front Pharmacol 2022; 13:958022. [PMID: 36176437 PMCID: PMC9514046 DOI: 10.3389/fphar.2022.958022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
It is worth noting that neuroinflammation is well recognized as a symptom of neurodegenerative diseases (NDs). The regulation of neuroinflammation becomes an attractive focus for innovative ND treatment technologies. There is evidence that IL-22 is associated with the development and progression of a wide assortment of NDs. For example, IL-22 can activate glial cells, causing them to generate pro-inflammatory cytokines and encourage lymphocyte infiltration in the brain. IL-22 mRNA is highly expressed in Alzheimer’s disease (AD) patients, and a high expression of IL-22 has also been detected in the brains of patients with other NDs. We examine the role of IL-22 in the development and treatment of NDs in this review, and we believe that IL-22 has therapeutic potential in these diseases.
Collapse
Affiliation(s)
- Wenjian Chen
- Department of Orthopaedics, Anhui Provincial Children’s Hospital, Hefei, China
| | - Jianpeng Wang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Huaizhi Yang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Yuankai Sun
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuchen Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanxun Han
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ming Shan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, China
- *Correspondence: Ming Shan, ; Junfeng Zhan,
| | - Junfeng Zhan
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ming Shan, ; Junfeng Zhan,
| |
Collapse
|
14
|
Noubissi Nzeteu GA, Gibbs BF, Kotnik N, Troja A, Bockhorn M, Meyer NH. Nanoparticle-based immunotherapy of pancreatic cancer. Front Mol Biosci 2022; 9:948898. [PMID: 36106025 PMCID: PMC9465485 DOI: 10.3389/fmolb.2022.948898] [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: 05/20/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) has a complex and unique tumor microenvironment (TME). Due to the physical barrier formed by the desmoplastic stroma, the delivery of drugs to the tumor tissue is limited. The TME also contributes to resistance to various immunotherapies such as cancer vaccines, chimeric antigen receptor T cell therapy and immune checkpoint inhibitors. Overcoming and/or modulating the TME is therefore one of the greatest challenges in developing new therapeutic strategies for PC. Nanoparticles have been successfully used as drug carriers and delivery systems in cancer therapy. Recent experimental and engineering developments in nanotechnology have resulted in increased drug delivery and improved immunotherapy for PC. In this review we discuss and analyze the current nanoparticle-based immunotherapy approaches that are at the verge of clinical application. Particularly, we focus on nanoparticle-based delivery systems that improve the effectiveness of PC immunotherapy. We also highlight current clinical research that will help to develop new therapeutic strategies for PC and especially targeted immunotherapies based on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Gaetan Aime Noubissi Nzeteu
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
- *Correspondence: N. Helge Meyer, ; Gaetan Aime Noubissi Nzeteu,
| | - Bernhard F. Gibbs
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Nika Kotnik
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
| | - Achim Troja
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - Maximilian Bockhorn
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
| | - N. Helge Meyer
- University Hospital of General and Visceral Surgery, Department of Human Medicine, University of Oldenburg and Klinikum Oldenburg, Oldenburg, Germany
- *Correspondence: N. Helge Meyer, ; Gaetan Aime Noubissi Nzeteu,
| |
Collapse
|
15
|
Chowdhury P, Bhusetty Nagesh PK, Hollingsworth TJ, Jaggi M, Chauhan SC, Yallapu MM. Coating a Self-Assembly Nanoconstruct with a Neutrophil Cell Membrane Enables High Specificity for Triple Negative Breast Cancer Treatment. ACS APPLIED BIO MATERIALS 2022; 5:4554-4566. [PMID: 35976626 DOI: 10.1021/acsabm.2c00614] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Breast cancer is one of the most commonly diagnosed cancers in American women. Triple negative breast cancer is among the most advanced and aggressive forms of breast cancer. Treatment options are limited for such cancers, making chemotherapy a convenient and effective treatment. Although these therapies can reduce morbidity and mortality, it is often followed by systemic side effects or relapse. Nanoparticles (NPs) have been considered for drug delivery approaches due to their ability to target various disease sites. Herein, we aim to develop a biomimetic NP construct (cell membrane-cloaked NPs) that exhibits specific affinity with triple negative breast cancer cells. In this regard, we designed biomimetic supramolecular nanoconstructs composed of a poly(vinyl pyrrolidone)-tannic acid (PVP-TA NPs/ PVT NPs) core and biofunctionalized with neutrophil cell membranes (PVT-NEU NPs). In this study, we have synthesized a PVT-NEU NP construct, characterized it, and evaluated it for improved targeting and therapeutic benefits in in vitro and in vivo models. Analysis of PVT-NEU NPs confirms the presence of the core of PVP-TA NPs coated with activated human neutrophil membranes. The study results confirmed that PVT-NEU NPs demonstrated an enhanced interaction and targeting with the tumor cells, thus improving the therapeutic activity of a model therapeutic agent (paclitaxel). Altogether, this study suggests the potential of biomimetic NPs as a promising therapeutic option for targeted drug delivery for advanced-stage breast cancer and other similar diseased conditions.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Prashanth Kumar Bhusetty Nagesh
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Laboratory of Signal Transduction, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - T J Hollingsworth
- Department of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Subhash Chand Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| | - Murali Mohan Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas 78504, United States
| |
Collapse
|
16
|
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
|
17
|
Khan MI, Zahra QUA, Batool F, Kalsoom F, Gao S, Ali R, Wang W, Kazmi A, Lianliang L, Wang G, Bilal M. Current Nano-Strategies to Improve Therapeutic Efficacy Across Special Structures. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100049] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Khedri M, Afsharchi F, Souderjani AH, Rezvantalab S, Didandeh M, Maleki R, Musaie K, Santos HA, Shahbazi M. Molecular scale study on the interactions of biocompatible nanoparticles with macrophage membrane and blood proteins. NANO SELECT 2022. [DOI: 10.1002/nano.202200043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Mohammad Khedri
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Fatemeh Afsharchi
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC) Zanjan University of Medical Sciences Zanjan Iran
| | - Amirhosein Hasanpour Souderjani
- Department of Pharmaceutical Engineering, School of Chemical Engineering College of Engineering, University of Tehran Tehran Iran
| | - Sima Rezvantalab
- Renewable Energies Department Faculty of Chemical Engineering Urmia University of Technology Urmia Iran
| | - Mohsen Didandeh
- Department of Chemical Engineering Tarbiat Modares University Tehran Iran
| | - Reza Maleki
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Kiyan Musaie
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC) Zanjan University of Medical Sciences Zanjan Iran
| | - Hélder A. Santos
- Department of Biomedical Engineering University Medical Center Groningen University of Groningen Groningen The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science University of Groningen/University Medical Center Groningen Groningen The Netherlands
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki Finland
| | - Mohammad‐Ali Shahbazi
- Department of Biomedical Engineering University Medical Center Groningen University of Groningen Groningen The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science University of Groningen/University Medical Center Groningen Groningen The Netherlands
| |
Collapse
|
19
|
Emerging Nanotherapeutic Approaches to Overcome Drug Resistance in Cancers with Update on Clinical Trials. Pharmaceutics 2022; 14:pharmaceutics14040866. [PMID: 35456698 PMCID: PMC9028322 DOI: 10.3390/pharmaceutics14040866] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
A key issue with modern cancer treatments is the emergence of resistance to conventional chemotherapy and molecularly targeted medicines. Cancer nanotherapeutics were created in order to overcome the inherent limitations of traditional chemotherapeutics. Over the last few decades, cancer nanotherapeutics provided unparalleled opportunities to understand and overcome drug resistance through clinical assessment of rationally designed nanoparticulate delivery systems. In this context, various design strategies such as passive targeting, active targeting, nano-drug, and multimodal nano-drug combination therapy provided effective cancer treatment. Even though cancer nanotherapy has made great technological progress, tumor biology complexity and heterogeneity and a lack of comprehensive knowledge of nano-bio interactions remain important roadblocks to future clinical translation and commercialization. The current developments and advancements in cancer nanotherapeutics employing a wide variety of nanomaterial-based platforms to overcome cancer treatment resistance are discussed in this article. There is also a review of various nanotherapeutics-based approaches to cancer therapy, including targeting strategies for the tumor microenvironment and its components, advanced delivery systems for specific targeting of cancer stem cells (CSC), as well as exosomes for delivery strategies, and an update on clinical trials. Finally, challenges and the future perspective of the cancer nanotherapeutics to reverse cancer drug resistance are discussed.
Collapse
|
20
|
Li Y, Chen Z, Gu L, Duan Z, Pan D, Xu Z, Gong Q, Li Y, Zhu H, Luo K. Anticancer nanomedicines harnessing tumor microenvironmental components. Expert Opin Drug Deliv 2022; 19:337-354. [PMID: 35244503 DOI: 10.1080/17425247.2022.2050211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Small-molecular drugs are extensively used in cancer therapy, while they have issues of nonspecific distribution and consequent side effects. Nanomedicines that incorporate chemotherapeutic drugs have been developed to enhance the therapeutic efficacy of these drugs and reduce their side effects. One of the promising strategies is to prepare nanomedicines by harnessing the unique tumor microenvironment (TME). AREAS COVERED The TME contains numerous cell types that specifically express specific antibodies on the surface including tumor vascular endothelial cells, tumor-associated adipocytes, tumor-associated fibroblasts, tumor-associated immune cells and cancer stem cells. The physicochemical environment is characterized with a low pH, hypoxia, and a high redox potential resulting from tumor-specific metabolism. The intelligent nanomedicines can be categorized into two groups: the first group which is rapidly responsive to extracellular chemical/biological factors in the TME and the second one which actively and/or specifically targets cellular components in the TME. EXPERT OPINION In this paper, we review recent progress of nanomedicines by harnessing the TME and illustrate the principles and advantages of different strategies for designing nanomedicines, which are of great significance for exploring novel nanomedicines or translating current nanomedicines into clinical practice. We will discuss the challenges and prospects of preparing nanomedicines to utilize or alter the TME for achieving effective, safe anticancer treatment.
Collapse
Affiliation(s)
- Yinggang Li
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhonglan Chen
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyu Duan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuping Xu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Youping Li
- Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
21
|
Nanomedicine for Immunotherapy Targeting Hematological Malignancies: Current Approaches and Perspective. NANOMATERIALS 2021; 11:nano11112792. [PMID: 34835555 PMCID: PMC8619332 DOI: 10.3390/nano11112792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/04/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022]
Abstract
Conventional chemotherapy has partial therapeutic effects against hematological malignancies and is correlated with serious side effects and great risk of relapse. Recently, immunotherapeutic drugs have provided encouraging results in the treatment of hematological malignancies. Several immunotherapeutic antibodies and cell therapeutics are in dynamic development such as immune checkpoint blockades and CAR-T treatment. However, numerous problems restrain the therapeutic effectiveness of tumor immunotherapy as an insufficient anti-tumor immune response, the interference of an immune-suppressive bone marrow, or tumoral milieu with the discharge of immunosuppressive components, access of myeloid-derived suppressor cells, monocyte intrusion, macrophage modifications, all factors facilitating the tumor to escape the anti-cancer immune response, finally reducing the efficiency of the immunotherapy. Nanotechnology can be employed to overcome each of these aspects, therefore having the possibility to successfully produce anti-cancer immune responses. Here, we review recent findings on the use of biomaterial-based nanoparticles in hematological malignancies immunotherapy. In the future, a deeper understanding of tumor immunology and of the implications of nanomedicine will allow nanoparticles to revolutionize tumor immunotherapy, and nanomedicine approaches will reveal their great potential for clinical translation.
Collapse
|
22
|
Tang L, Mei Y, Shen Y, He S, Xiao Q, Yin Y, Xu Y, Shao J, Wang W, Cai Z. Nanoparticle-Mediated Targeted Drug Delivery to Remodel Tumor Microenvironment for Cancer Therapy. Int J Nanomedicine 2021; 16:5811-5829. [PMID: 34471353 PMCID: PMC8403563 DOI: 10.2147/ijn.s321416] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/14/2021] [Indexed: 12/24/2022] Open
Abstract
Advanced research has revealed the crucial role of tumor microenvironment (TME) in tumorigenesis. TME consists of a complicated network with a variety of cell types including endothelial cells, pericytes, immune cells, cancer-associated fibroblasts (CAFs), cancer stem cells (CSCs) as well as the extracellular matrix (ECM). The TME-constituting cells interact with the cancerous cells through plenty of signaling mechanisms and pathways in a dynamical way, participating in tumor initiation, progression, metastasis, and response to therapies. Hence, TME is becoming an attractive therapeutic target in cancer treatment, exhibiting potential research interest and clinical benefits. Presently, the novel nanotechnology applied in TME regulation has made huge progress. The nanoparticles (NPs) can be designed as demand to precisely target TME components and to inhibit tumor progression through TME modulation. Moreover, nanotechnology-mediated drug delivery possesses many advantages including prolonged circulation time, enhanced bioavailability and decreased toxicity over traditional therapeutic modality. In this review, update information on TME remodeling through NPs-based targeted drug delivery strategies for anticancer therapy is summarized.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yan Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Qiaqia Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yonggang Xu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jie Shao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Zihao Cai
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
23
|
Gao S, Yang X, Xu J, Qiu N, Zhai G. Nanotechnology for Boosting Cancer Immunotherapy and Remodeling Tumor Microenvironment: The Horizons in Cancer Treatment. ACS NANO 2021; 15:12567-12603. [PMID: 34339170 DOI: 10.1021/acsnano.1c02103] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Immunotherapy that harnesses the human immune system to fight cancer has received widespread attention and become a mainstream strategy for cancer treatment. Cancer immunotherapy not only eliminates primary tumors but also treats metastasis and recurrence, representing a major advantage over traditional cancer treatments. Recently with the development of nanotechnology, there exists much work applying nanomaterials to cancer immunotherapy on the basis of their excellent physiochemical properties, such as efficient tissue-specific delivery function, huge specific surface area, and controllable surface chemistry. Consequently, nanotechnology holds significant potential in improving the efficacy of cancer immunotherapy. Nanotechnology-based immunotherapy mainly manifests its inhibitory effect on tumors via two different approaches: one is to produce an effective anti-tumor immune response during tumorigenesis, and the other is to enhance tumor immune defense ability by modulating the immune suppression mechanism in the tumor microenvironment. With the success of tumor immunotherapy, understanding the interaction between the immune system and smart nanomedicine has provided vigorous vitality for the development of cancer treatment. This review highlights the application, progress, and prospect of nanomedicine in the process of tumor immunoediting and also discusses several engineering methods to improve the efficiency of tumor treatment.
Collapse
Affiliation(s)
- Shan Gao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Xiaoye Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Jiangkang Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Na Qiu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| | - Guangxi Zhai
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Shandong University, 44 WenhuaXilu, Jinan 250012, China
| |
Collapse
|
24
|
Yong HW, Kakkar A. Nanoengineering Branched Star Polymer-Based Formulations: Scope, Strategies, and Advances. Macromol Biosci 2021; 21:e2100105. [PMID: 34117840 DOI: 10.1002/mabi.202100105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/26/2021] [Indexed: 12/24/2022]
Abstract
Soft nanoparticles continue to offer a promising platform for the encapsulation and controlled delivery of poorly water-soluble drugs and help enhance their bioavailability at targeted sites. Linear amphiphilic block copolymers are the most extensively investigated in formulating delivery vehicles. However, more recently, there has been increasing interest in utilizing branched macromolecules for nanomedicine, as these have been shown to lower critical micelle concentrations, form particles of smaller dimensions, facilitate the inclusion of varied compositions and function-based entities, as well as provide prolonged and sustained release of cargo. In this review, it is aimed to discuss some of the key variables that are studied in tailoring branched architecture-based assemblies, and their influence on drug loading and delivery. By understanding structure-property relationships in these formulations, one can better design branched star polymers with suitable characteristics for efficient therapeutic interventions. The role played by polymer composition, chain architecture, crosslinking, stereocomplexation, compatibility between polymers and drugs, drug/polymer concentrations, and self-assembly methods in their performance as nanocarriers is highlighted.
Collapse
Affiliation(s)
- Hui Wen Yong
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Quebec, H3A 0B8, Canada
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Quebec, H3A 0B8, Canada
| |
Collapse
|
25
|
Biomarkers in Pancreatic Cancer as Analytic Targets for Nanomediated Imaging and Therapy. MATERIALS 2021; 14:ma14113083. [PMID: 34199998 PMCID: PMC8200189 DOI: 10.3390/ma14113083] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022]
Abstract
As the increase in therapeutic and imaging technologies is swiftly improving survival chances for cancer patients, pancreatic cancer (PC) still has a grim prognosis and a rising incidence. Practically everything distinguishing for this type of malignancy makes it challenging to treat: no approved method for early detection, extended asymptomatic state, limited treatment options, poor chemotherapy response and dense tumor stroma that impedes drug delivery. We provide a narrative review of our main findings in the field of nanoparticle directed treatment for PC, with a focus on biomarker targeted delivery. By reducing drug toxicity, increasing their tumor accumulation, ability to modulate tumor microenvironment and even improve imaging contrast, it seems that nanotechnology may one day give hope for better outcome in pancreatic cancer. Further conjugating nanoparticles with biomarkers that are overexpressed amplifies the benefits mentioned, with potential increase in survival and treatment response.
Collapse
|
26
|
Mushtaq A, Li L, A A, Grøndahl L. Chitosan Nanomedicine in Cancer Therapy: Targeted Delivery and Cellular Uptake. Macromol Biosci 2021; 21:e2100005. [PMID: 33738977 DOI: 10.1002/mabi.202100005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/22/2021] [Indexed: 12/11/2022]
Abstract
Nanomedicine has gained much attention for the management and treatment of cancers due to the distinctive physicochemical properties of the drug-loaded particles. Chitosan's cationic nature is attractive for the development of such particles for drug delivery, transfection, and controlled release. The particle properties can be improved by modification of the polymer or the particle themselves. The physicochemical properties of chitosan particles are analyzed in 126 recent studies, which allows to highlight their impact on passive and active targeted drug delivery, cellular uptake, and tumor growth inhibition (TGI). From 2012 to 2019, out of 40 in vivo studies, only 4 studies are found reporting a reduction in tumor size by using chitosan particles while all other studies reported tumor growth inhibition relative to controls. A total of 23 studies are analyzed for cellular uptake including 12 studies reporting cellular uptake mechanisms. Understanding and exploiting the processes involved in targeted delivery, endocytosis, and exocytosis by controlling the physicochemical properties of chitosan particles are important for the development of safe and efficient nanomedicine. It is concluded based on the recent literature available on chitosan particles that combination therapies can play a pivotal role in transformation of chitosan nanomedicine from bench to bedside.
Collapse
Affiliation(s)
- Asim Mushtaq
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Li Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Anitha A
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia
| | - Lisbeth Grøndahl
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 68, Cooper Road, Brisbane, Queensland, 4072, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Building 75, Corner of College and Cooper Road, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
27
|
Choi W, Battistella C, Gianneschi NC. High efficiency loading of micellar nanoparticles with a light switch for enzyme-induced rapid release of cargo. Biomater Sci 2021; 9:653-657. [PMID: 33300507 PMCID: PMC9753762 DOI: 10.1039/d0bm01713b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Polymeric nanoscale materials able to target and accumulate in the tumor microenvironment (TME) offer promising routes for a safer delivery of anticancer drugs. By reaching their targets before significant amounts of drug are released, such materials can reduce off-target side effects and maximize drug concentration in the TME. However, poor drug loading capacity and inefficient nanomaterial penetration into the tumor can limit their therapeutic efficacy. Herein, we provide a novel approach to achieve high loading profiles while ensuring fast and efficient drug penetration in the tumor. This is achieved by co-polymerizing light-sensitive paclitaxel with monomers responsive to tumor-associated enzymes, and assembling the resulting di-block copolymers into spherical micelles. While light exposure enables paclitaxel to decouple from the polymeric backbone into light-activated micelles, enzymatic digestion in the TME initiates its burst release. Through a series of in vitro cytotoxicity assays, we demonstrate that these light-switch micelles hold greater potency than covalently linked, non-triggered micelles, and enable therapeutic profiles comparable to that of the free drug.
Collapse
Affiliation(s)
- Wonmin Choi
- Department of Chemistry, Department of Materials Science & Engineering, Department of Biomedical Engineering, Department of Pharmacology, International Institute for Nanotechnology, Simpson-Querrey Institute, Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208, USA.
| | | | | |
Collapse
|
28
|
Molecular targeted treatment and drug delivery system for gastric cancer. J Cancer Res Clin Oncol 2021; 147:973-986. [PMID: 33550445 DOI: 10.1007/s00432-021-03520-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/10/2021] [Indexed: 12/24/2022]
Abstract
Gastric cancer is still a major cancer worldwide. The early diagnosis rate of gastric cancer in most high incidence countries is low. At present, the overall treatment effect of gastric cancer is poor, and the median overall survival remains low. Most of the patients with gastric cancer are in an advanced stage when diagnosed, and drug treatment has become the main means. Thus, new targeted drugs and therapeutic strategies are the hope of improving the therapeutic effect of gastric cancer. In this review, we summarize the new methods and advances of targeted therapy for gastric cancer, including novel molecular targeted therapeutic agents and drug delivery systems, with a major focus on the development of drug delivery systems (drug carriers and targeting peptides). Elaborating these new methods and advances will contribute to the management of gastric cancer.
Collapse
|
29
|
Yang M, Li J, Gu P, Fan X. The application of nanoparticles in cancer immunotherapy: Targeting tumor microenvironment. Bioact Mater 2020; 6:1973-1987. [PMID: 33426371 PMCID: PMC7773537 DOI: 10.1016/j.bioactmat.2020.12.010] [Citation(s) in RCA: 316] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/04/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor development and metastasis are closely related to the structure and function of the tumor microenvironment (TME). Recently, TME modulation strategies have attracted much attention in cancer immunotherapy. Despite the preliminary success of immunotherapeutic agents, their therapeutic effects have been restricted by the limited retention time of drugs in TME. Compared with traditional delivery systems, nanoparticles with unique physical properties and elaborate design can efficiently penetrate TME and specifically deliver to the major components in TME. In this review, we briefly introduce the substitutes of TME including dendritic cells, macrophages, fibroblasts, tumor vasculature, tumor-draining lymph nodes and hypoxic state, then review various nanoparticles targeting these components and their applications in tumor therapy. In addition, nanoparticles could be combined with other therapies, including chemotherapy, radiotherapy, and photodynamic therapy, however, the nanoplatform delivery system may not be effective in all types of tumors due to the heterogeneity of different tumors and individuals. The changes of TME at various stages during tumor development are required to be further elucidated so that more individualized nanoplatforms could be designed.
Collapse
Key Words
- AC-NPs, antigen-capturing nanoparticles
- ANG2, angiopoietin-2
- APCs, antigen-presenting cells
- Ab, antibodies
- Ag, antigen
- AuNCs, gold nanocages
- AuNPs, gold nanoparticles
- BBB, blood-brain barrier
- BTK, Bruton's tyrosine kinase
- Bcl-2, B-cell lymphoma 2
- CAFs, cancer associated fibroblasts
- CAP, cleavable amphiphilic peptide
- CAR-T, Chimeric antigen receptor-modified T-cell therapy
- CCL, chemoattractant chemokines ligand
- CTL, cytotoxic T lymphocytes
- CTLA4, cytotoxic lymphocyte antigen 4
- CaCO3, calcium carbonate
- Cancer immunotherapy
- DCs, dendritic cells
- DMMA, 2,3-dimethylmaleic anhydrid
- DMXAA, 5,6-dimethylxanthenone-4-acetic acid
- DSF/Cu, disulfiram/copper
- ECM, extracellular matrix
- EGFR, epidermal growth factor receptor
- EMT, epithelial-mesenchymal transition
- EPG, egg phosphatidylglycerol
- EPR, enhanced permeability and retention
- FAP, fibroblast activation protein
- FDA, the Food and Drug Administration
- HA, hyaluronic acid
- HB-GFs, heparin-binding growth factors
- HIF, hypoxia-inducible factor
- HPMA, N-(2-hydroxypropyl) methacrylamide
- HSA, human serum albumin
- Hypoxia
- IBR, Ibrutinib
- IFN-γ, interferon-γ
- IFP, interstitial fluid pressure
- IL, interleukin
- LMWH, low molecular weight heparin
- LPS, lipopolysaccharide
- M2NP, M2-like TAM dual-targeting nanoparticle
- MCMC, mannosylated carboxymethyl chitosan
- MDSCs, myeloid-derived suppressor cells
- MPs, microparticles
- MnO2, manganese dioxide
- NF-κB, nuclear factor κB
- NK, nature killer
- NO, nitric oxide
- NPs, nanoparticles
- Nanoparticles
- ODN, oligodeoxynucleotides
- PD-1, programmed cell death protein 1
- PDT, photodynamic therapy
- PFC, perfluorocarbon
- PHDs, prolyl hydroxylases
- PLGA, poly(lactic-co-glycolic acid)
- PS, photosensitizer
- PSCs, pancreatic stellate cells
- PTX, paclitaxel
- RBC, red-blood-cell
- RLX, relaxin-2
- ROS, reactive oxygen species
- SA, sialic acid
- SPARC, secreted protein acidic and rich in cysteine
- TAAs, tumor-associated antigens
- TAMs, tumor-associated macrophages
- TDPA, tumor-derived protein antigens
- TGF-β, transforming growth factor β
- TIE2, tyrosine kinase with immunoglobulin and epidermal growth factor homology domain 2
- TIM-3, T cell immunoglobulin domain and mucin domain-3
- TLR, Toll-like receptor
- TME, tumor microenvironment
- TNF-α, tumor necrosis factor alpha
- TfR, transferrin receptor
- Tregs, regulatory T cells
- Tumor microenvironment
- UPS-NP, ultra-pH-sensitive nanoparticle
- VDA, vasculature disrupting agent
- VEGF, vascular endothelial growth factor
- cDCs, conventional dendritic cells
- melittin-NP, melittin-lipid nanoparticle
- nMOFs, nanoscale metal-organic frameworks
- scFv, single-chain variable fragment
- siRNA, small interfering RNA
- tdLNs, tumor-draining lymph nodes
- α-SMA, alpha-smooth muscle actin
Collapse
|
30
|
Localized delivery of active targeting micelles from nanofibers patch for effective breast cancer therapy. Int J Pharm 2020; 584:119412. [PMID: 32418898 DOI: 10.1016/j.ijpharm.2020.119412] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 01/08/2023]
Abstract
Nanofibers based transdermal drug delivery is a promising platform, and it effectively delivers the drug to tumor sites. The objective of the study was to fabricate stimuli-responsive polymeric nanofibers encapsulated with an active targeting micellar system for in situ drug delivery. Stimuli-responsive core-shell nanofibers release thedrug at target sites with minimum side effects to the other organs, decrease the drug administration concentration. Initially, we prepared CA conjugated PCPP polymeric micelles loaded with PTX. Then, core-shell nanofibers were prepared using PHM with coaxial electrospinning and distinct core-shell nanofibers formation confirm by SEM and TEM. Nanofibers showed a homogenous distribution of micelles inside the fiber mesh, diffusion, and erosion processes lead to a controlled release of PTX.In vitro drug release and swelling, revealed the pH based sustained release of the drug for 180 h from the nanofibers mat. Functional and stimuli-responsive nanofibers highly absorb H+ ions and repulsion of cations promoting maximum swelling to release more drugs in acidic pH. An increased transportation rate of 70% drug release through epidermis for 120 h. Nanofibers effectively internalize to the skin, and it confirmed by confocal microscopy. MCF-7 cells grown and spread over the nanofibers, which show the biocompatibility of nanofibers. Compared to PTX, drug-loaded nanofibers exhibited higher cytotoxicity for 8 days which was confirmed by the flow cytometry. These promising results confirm, the novel stimuli-responsive core-shell nanofibers actively target breast cancer cells and lead the way to safe cancer therapy.
Collapse
|
31
|
Wang J, Wu X, Shen P, Wang J, Shen Y, Shen Y, Webster TJ, Deng J. Applications of Inorganic Nanomaterials in Photothermal Therapy Based on Combinational Cancer Treatment. Int J Nanomedicine 2020; 15:1903-1914. [PMID: 32256067 PMCID: PMC7094149 DOI: 10.2147/ijn.s239751] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/16/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cancer is one of the major causes of death and is difficult to cure using existing clinical therapies. Clinical cancer treatments [such as surgery, chemotherapy (CHT), radiotherapy (RT) and immunotherapy (IT)] are widely used but they have limited therapeutic effects and unavoidable side effects. Recently, the development of novel nanomaterials offers a platform for combinational therapy (meaning a combination of two or more therapeutic agents) which is a promising approach for cancer therapy. Recent studies have demonstrated several types of nanomaterials suitable for photothermal therapy (PTT) based on a near-infrared (NIR) light-responsive system. PTT possesses favorable properties such as being low in cost, and having high temporospatial control with minimal invasiveness. However, short NIR light penetration depth limits its functions. METHODS In this review, due to their promise, we focus on inorganic nanomaterials [such as hollow mesoporous silica nanoparticles (HMSNs), tungsten sulfide quantum dots (WS2QDs), and gold nanorods (AuNRs)] combining PTT with CHT, RT or IT in one treatment, aiming to provide a comprehensive understanding of PTT-based combinational cancer therapy. RESULTS This review found much evidence for the use of inorganic nanoparticles for PTT-based combinational cancer therapy. CONCLUSION Under synergistic effects, inorganic nanomaterial-based combinational treatments exhibit enhanced therapeutic effects compared to PTT, CHT, RT, IT or PDT alone and should be further investigated in the cancer field.
Collapse
Affiliation(s)
- Ji Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Xia Wu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Peng Shen
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, People’s Republic of China
| | - Jun Wang
- Department of General Surgery, The Fifth People’s Hospital of Wujiang, Suzhou, People’s Republic of China
| | - Yidan Shen
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yan Shen
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, People’s Republic of China
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Junjie Deng
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, People’s Republic of China
| |
Collapse
|
32
|
Abstract
This review highlights the pharmacokinetic features and tumor imaging preponderance of renal clearable AuNCs for in vivo tumor imaging.
Collapse
Affiliation(s)
- Huili Li
- Engineering Research Center of Cell and Therapeutic Antibody
- Ministry of Education
- School of Pharmacy
- Shanghai Jiaotong University
- Shanghai 200240
| | - Hongle Li
- Department of Molecular Pathology
- The Affiliated Cancer Hospital
- Zhengzhou University
- Zhengzhou
- China
| | - Ajun Wan
- National Engineering Research Center of Protected Agriculture
- School of Medicine
- Tongji University
- Shanghai 200092
- China
| |
Collapse
|
33
|
Chen M, Xu G, Fan M, Jia H, Xiao L, Lang J. Anti-tumour effects of a xenogeneic fibroblast activation protein-based whole cell tumour vaccine in murine tumour models. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:4182-4193. [PMID: 31722575 DOI: 10.1080/21691401.2019.1687498] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The clinical benefit of cancer immunotherapy, including tumour vaccines, is influenced by immunosuppressive factors in the tumour microenvironment. Among these factors, cancer-associated fibroblasts (CAFs) and their products, such as fibroblast activation protein-α (FAPα), greatly affect tumourigenesis, development, metastasis and treatment tolerance, which make them promising immunotherapy targets for cancer patients. Our previous study reported that a whole cell tumour vaccine (WCTV) expressing FAPα inhibited tumour growth by simultaneously attacking cancer cells and CAFs. This study aimed to improve WCTVs with xenoantigens to end immune tolerance and to further activate the adaptive immune system. In the present study, we designed a WCTV by transducing a vector encoding human FAPα (hFAPα) into murine tumour cells and evaluated its efficacy in multiple solid tumour models. Immunotherapy with this WCTV effectively delayed tumour growth and prevented recurrence. The anti-tumour responses were clearly linked to antigen-specific cytotoxic T cells, whereas CD4(+) T lymphocytes also played a role. Humoural immune responses were activated because the adoptive transfer of immunoglobulins induced abscopal anti-tumour effects, and autoantibodies against FAPα were specifically detected in the sera of immunized mice. Moreover, an increased number of apoptotic tumour cells along with a reduced number of CAFs within the tumours suggest that xenogeneic FAPα-based WCTV has the potential to drive T cell and antibody responses against cancer cells and CAFs. This finding could offer an advanced strategy to treat multiple solid tumours with individualized cancer immunotherapy techniques.
Collapse
Affiliation(s)
- Meihua Chen
- Department of Radiation Oncology, School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.,Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Guangchao Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ming Fan
- Department of Radiation Oncology, School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.,Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Hongyuan Jia
- Department of Radiation Oncology, School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.,Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Ling Xiao
- Department of Radiation Oncology, School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.,Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| | - Jinyi Lang
- Department of Radiation Oncology, School of Medicine, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.,Radiation Oncology Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
34
|
Cell-Penetrating Peptide and Transferrin Co-Modified Liposomes for Targeted Therapy of Glioma. Molecules 2019; 24:molecules24193540. [PMID: 31574945 PMCID: PMC6804123 DOI: 10.3390/molecules24193540] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/20/2019] [Accepted: 09/25/2019] [Indexed: 12/02/2022] Open
Abstract
Glioma is one of the most aggressive and common malignant brain tumors. Due to the presence of the blood-brain barrier (BBB), the effectiveness of therapeutics is greatly affected. In this work, to develop an efficient anti-glioma drug with targeting and which was able to cross the BBB, cell-penetrating peptides (R8) and transferrin co-modified doxorubicin (DOX)-loaded liposomes (Tf-LPs) were prepared. Tf-LPs possessed a spherical shape and uniform size with 128.64 nm and their ξ-potential was 6.81 mV. Tf-LPs were found to be stable in serum within 48 h. Uptake of Tf-LPs in both U87 and GL261 cells was analyzed by confocal laser scanning microscopy and by flow cytometry. Tf-LPs were efficiently taken up by both U87 and GL261 cells. Moreover, Tf-LPs exhibited sustained-release. The cumulative release of DOX from Tf-LPs reached ~50.0% and showed excellent anti-glioma efficacy. Histology of major organs, including brain, heart, liver, spleen, lungs and kidney, and the bodyweight of mice, all indicated low toxicity of Tf-LPs. In conclusion, Tf-LPs showed great promise as an anti-glioma therapeutic agent.
Collapse
|
35
|
Gao J, Fan K, Jin Y, Zhao L, Wang Q, Tang Y, Xu H, Liu Z, Wang S, Lin J, Lin D. PEGylated lipid bilayer coated mesoporous silica nanoparticles co-delivery of paclitaxel and curcumin leads to increased tumor site drug accumulation and reduced tumor burden. Eur J Pharm Sci 2019; 140:105070. [PMID: 31518679 DOI: 10.1016/j.ejps.2019.105070] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 09/07/2019] [Accepted: 09/09/2019] [Indexed: 01/13/2023]
Abstract
Homogeneous PEGylated lipid bilayer coated highly ordered MSNs (PLMSNs) which were systematically optimized and characterized to co-encapsulate paclitaxel (Tax) and curcumin (Cur) were verified to manifest prolonged and enhanced cytotoxic effect against canine breast cancer cells in our previous study. In this article, we took further study of the pharmacokinetic property, cellular uptake, subcellular localization, in vivo distribution and tumor accumulation ability, and treatment efficacy of the drug delivery system. The results revealed that the delivery system could significantly increase the AUC of two drugs, and the anti-tumor effect showed that both intravenous and intratumoral administration group better controlled the tumor weight than that of other groups (P < .05), and the anti-tumor rates were 58.4% and 58.3% respectively. Cell uptake and localization study showed that PLMSNs could effectively carry drugs into cancer cells with sustained release characteristics. The subcellular localization of PLMSNs was mainly in lysosomes and mitochondria. In vivo fluorescence tracing results showed that PLMSNs could be effectively accumulated in the tumor site. The results revealed that the delivery system could effectively reduce the clinical dosage of drugs and reduce its toxic side effects, effectively carry drugs into cancer cells, and exhibit good targeting characteristics for breast cancer.
Collapse
Affiliation(s)
- Jiafeng Gao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Kai Fan
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Yipeng Jin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Linna Zhao
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Qian Wang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Yinian Tang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Huihao Xu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Zhongjie Liu
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Shuaiyu Wang
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China
| | - Jiahao Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China.
| | - Degui Lin
- The Clinical Department, College of Veterinary Medicine, China Agricultural University, Beijing, PR China.
| |
Collapse
|
36
|
Hu G, Guo M, Xu J, Wu F, Fan J, Huang Q, Yang G, Lv Z, Wang X, Jin Y. Nanoparticles Targeting Macrophages as Potential Clinical Therapeutic Agents Against Cancer and Inflammation. Front Immunol 2019; 10:1998. [PMID: 31497026 PMCID: PMC6712945 DOI: 10.3389/fimmu.2019.01998] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
With the development of nanotechnology, significant progress has been made in the design, and manufacture of nanoparticles (NPs) for use in clinical treatments. Recent increases in our understanding of the central role of macrophages in the context of inflammation and cancer have reinvigorated interest in macrophages as drug targets. Macrophages play an integral role in maintaining the steady state of the immune system and are involved in cancer and inflammation processes. Thus, NPs tailored to accurately target macrophages have the potential to transform disease treatment. Herein, we first present a brief background information of NPs as drug carriers, including but not limited to the types of nanomaterials, their biological properties and their advantages in clinical application. Then, macrophage effector mechanisms and recent NPs-based strategies aimed at targeting macrophages by eliminating or re-educating macrophages in inflammation and cancer are summarized. Additionally, the development of nanocarriers targeting macrophages for disease diagnosis is also discussed. Finally, the significance of macrophage-targeting nanomedicine is highlighted, with the goal of facilitating future clinical translation.
Collapse
Affiliation(s)
- Guorong Hu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Mengfei Guo
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Juanjuan Xu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wu
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jinshuo Fan
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Huang
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhilei Lv
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Jin
- Key Laboratory of Respiratory Diseases of the Ministry of Health, Department of Respiratory and Critical Care Medicine, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Pathade AD, Kommineni N, Bulbake U, Thummar MM, Samanthula G, Khan W. Preparation and Comparison of Oral Bioavailability for Different Nano-formulations of Olaparib. AAPS PharmSciTech 2019; 20:276. [PMID: 31388783 DOI: 10.1208/s12249-019-1468-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/01/2019] [Indexed: 12/15/2022] Open
Abstract
Olaparib (OLA) is a poly ADP ribose polymerase (PARP) inhibitor approved for germline BRCA-mutated (gBRCAm) advanced ovarian cancer and breast cancer. Low oral bioavailability of this drug requires increase in the dose and frequency causing haematological toxicity in the patients. The purpose of this study is to prepare different nano-formulations of OLA lipospheres (LP) by melt dispersion and nano-suspensions (NSP) by solvent evaporation (SE) and wet milling (WM) techniques and compare oral bioavailability of these formulations. Size of the nano-formulations OLA-LP, OLA-NSPSE and OLA-NSPWM were found to be 126.71 ± 4.54, 128.6 ± 2.34 and 531.1 ± 5.34 nm with polydispersity index below 0.3. In vitro release studies were performed by dialysis bag method where the sustained drug release was observed from nano-formulations until 9 h with Higuchi for OLA suspended in 2.5% w/v sodium carboxy methyl cellulose (OLA-SP), OLA-LP and OLA-NSPWM and Peppas for OLA-NSPSE-based drug release kinetics. In vivo pharmacokinetic studies, haematological toxicity and distribution studies were performed on rats. Results showed that there was an improvement in Cmax, AUCtotal, t1/2 and MRT by OLA nano-formulations when compared with OLA-SP. OLA-SP has shown reduction in WBC, platelets and lymphocytes at 12 and 36 h time points; however, no reduction in cell count was observed with OLA nano-formulations. Distribution studies proved FITC nano-formulations were most rapidly absorbed and distributed when compared with FITC-loaded suspension. From the above results, it was concluded that OLA nano-formulations can be an alternative to enhance the oral bioavailability and to reduce the haematological toxicity of OLA.
Collapse
|
38
|
Gong BS, Wang R, Xu HX, Miao MY, Yao ZZ. Nanotherapy Targeting the Tumor Microenvironment. Curr Cancer Drug Targets 2019; 19:525-533. [DOI: 10.2174/1568009619666181220103714] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/11/2018] [Accepted: 11/13/2018] [Indexed: 01/24/2023]
Abstract
Cancer is characterized by high mortality and low curability. Recent studies have shown
that the mechanism of tumor resistance involves not only endogenous changes to tumor cells, but
also to the tumor microenvironment (TME), which provides the necessary conditions for the growth,
invasion, and metastasis of cancer cells, akin to Stephen Paget’s hypothesis of “seed and soil.”
Hence, the TME is a significant target for cancer therapy via nanoparticles, which can carry different
kinds of drugs targeting different types or stages of tumors. The key step of nanotherapy is the
achievement of accurate active or passive targeting to trigger drugs precisely at tumor cells, with less
toxicity and fewer side effects. With deepened understanding of the tumor microenvironment and
rapid development of the nanomaterial industry, the mechanisms of nanotherapy could be individualized
according to the specific TME characteristics, including low pH, cancer-associated fibroblasts
(CAFs), and increased expression of metalloproteinase. However, some abnormal features of the
TME limit drugs from reaching all tumor cells in lethal concentrations, and the characteristics of tumors
vary in numerous ways, resulting in great challenges for the clinical application of nanotherapy.
In this review, we discuss the essential role of the tumor microenvironment in the genesis and
development of tumors, as well as the measures required to improve the therapeutic effects of tumor
microenvironment-targeting nanoparticles and ways to reduce damage to normal tissue.
Collapse
Affiliation(s)
- Bo-Shen Gong
- Administration Office for Undergraduates, Second Military Medical University, Shanghai, 200433, China
| | - Rui Wang
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Hong-Xia Xu
- Department of Nutrition, Daping Hospital and Research Institute of Surgery, Third Military Medical University, Chongqing, 400042, China
| | - Ming-Yong Miao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| | - Zhen-Zhen Yao
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai, 200433, China
| |
Collapse
|
39
|
Kommineni N, Mahira S, Domb AJ, Khan W. Cabazitaxel-Loaded Nanocarriers for Cancer Therapy with Reduced Side Effects. Pharmaceutics 2019; 11:pharmaceutics11030141. [PMID: 30934535 PMCID: PMC6470818 DOI: 10.3390/pharmaceutics11030141] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023] Open
Abstract
Jevtana® is a micellar cabazitaxel (CBZ) solution that was approved for prostate cancer in 2010, and recently, this drug has been reported for breast cancer. The purpose of this study is to evaluate the mediated delivery of CBZ via liposomes and nanoparticles (NPs) for the treatment of breast cancer and compare these with a micellar formulation that is currently in clinical use. CBZ-loaded nanocarriers were prepared with particle sizes between 70–110 nm, and with the sustained in vitro release of CBZ for more than 28 days. Cytotoxicity studies on MCF-7 and MDA-MB-231 cells demonstrated the toxic potential of these nanocarriers. Cellular internalization revealed that NPs and liposomes have better permeability than micelles. Cell cycle analysis and apoptosis studies on MCF-7 and MDA-MB-231 cells confirmed G2/M phase arrest as well as cell death due to apoptosis and necrosis, where formulations were found to be effective compared to a micellar CBZ solution. Results from pharmacokinetic studies revealed that there is an increased circulation half-life and mean residence time for CBZ liposomes and NPs in comparison with a micellar CBZ solution. CBZ liposomes and NPs showed a reduction in hemolysis and neutropenia in comparison with a micellar CBZ solution in rats.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad 500037, India.
| | - Shaheen Mahira
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad 500037, India.
| | - Abraham J Domb
- School of Pharmacy-Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
40
|
Cai G, Wang S, Zhao L, Sun Y, Yang D, Lee RJ, Zhao M, Zhang H, Zhou Y. Thiophene Derivatives as Anticancer Agents and Their Delivery to Tumor Cells Using Albumin Nanoparticles. Molecules 2019; 24:E192. [PMID: 30621360 PMCID: PMC6337126 DOI: 10.3390/molecules24010192] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/27/2018] [Accepted: 12/29/2018] [Indexed: 01/25/2023] Open
Abstract
A series of thiophene derivatives (TPs) were synthesized and evaluated for cytotoxicity in HepG2 and SMMC-7721 cell lines by MTT assay. TP 5 was identified as a potential anticancer agent based on its ability to inhibit tumor cell growth. Drawbacks of TPs, including poor solubility and high toxicity, were overcome through delivery using self-assembling HSA nanoparticles (NPs). The optimum conditions for TP 5-NPs synthesis obtained by adjusting the temperature and concentration of TP 5. The NPs had an encapsulation efficiency of 99.59% and drug-loading capacity of 3.70%. TP 5 was slowly released from TP 5-NPs in vitro over 120 h. HepG2 and SMMC-7721 cell lines were employed to study cytotoxicity of TP 5-NPs, which exhibited high potency. ROS levels were elevated and mitochondrial membrane potentials reversed when the two cell lines were treated with TP 5-NPs for 12 h. Cellular uptake of fluorescence-labeled TP 5-NPs in vitro was analyzed by flow cytometry and laser confocal scanning microscopy. Fluorescence intensity increased over time, suggesting that TP 5-NPs were efficiently taken up by tumor cells. In conclusion, TP 5-NPs showed great promise as an anticancer therapeutic agent.
Collapse
Affiliation(s)
- Guangsheng Cai
- College of Life Sciences, Jilin University, Changchun 130012, China.
| | - Simiao Wang
- College of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lang Zhao
- College of Chemistry, Jilin University, Changchun 130012, China.
| | - Yating Sun
- College of Life Sciences, Jilin University, Changchun 130012, China.
| | - Dongsheng Yang
- School of Pharmaceutical and Food Sciences, Zhuhai College of Jilin University, Zhuhai 519041, China.
| | - Robert J Lee
- College of Life Sciences, Jilin University, Changchun 130012, China.
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| | - Menghui Zhao
- College of Life Sciences, Jilin University, Changchun 130012, China.
| | - Huan Zhang
- College of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yulin Zhou
- College of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
41
|
Wang Y, Wang B, Xiao S, Li Y, Chen Q. miR-125a/b inhibits tumor-associated macrophages mediated in cancer stem cells of hepatocellular carcinoma by targeting CD90. J Cell Biochem 2018; 120:3046-3055. [PMID: 30536969 DOI: 10.1002/jcb.27436] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022]
Abstract
Cancer stem cells promote tumorigenesis and progression of hepatocellular carcinoma (HCC). Recently, emerging evidence indicates tumor-associated macrophages (TAMs) play an important role in tumor progression. However, TAMs often occurs with unknown mechanisms. As an important mediator in intercellular communications, exosomes secreted by host cells mediate the exchange of genetic materials and proteins, which involves tumor aggressiveness. The aim of the study was to investigate whether exosomes derived from TAMs mediate stem cell properties in HCC. TAMs were isolated from the tissues of HCC. microRNA (miRNA) expression profiles of TAMs were analyzed using miRNA microarray. In vitro cell coculture was further conducted to investigate the crosstalk between TAMs and tumor cells mediated by TAMs exosomes. In this study, we showed that TAMs exosomes promote HCC cell proliferation and stem cell properties. Using miRNA profiles assay, we identified significantly lower levels of miR-125a and miR-125b in exosomes and cell lysate isolated from TAMs. Functional studies revealed that the HCC cells were treated with TAM exosomes or transfected with miR-125a/b suppressed cell proliferation and stem cell properties by targeting CD90, a stem cell marker of HCC stem cells. The study indicated that miR-125a/b targeting CD90 played important roles in cancer stem cells of HCC.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of General Surgery, Shanghai Tongji Hospital, Medical School of Tongji University, Shanghai, China
| | - Bingyi Wang
- Department of General Surgery, Shanghai Tongji Hospital, Medical School of Tongji University, Shanghai, China
| | - Shuai Xiao
- Department of General Surgery, Shanghai Tongji Hospital, Medical School of Tongji University, Shanghai, China
| | - Yang Li
- Department of General Surgery, Shanghai Tongji Hospital, Medical School of Tongji University, Shanghai, China
| | - Quanning Chen
- Department of General Surgery, Shanghai Tongji Hospital, Medical School of Tongji University, Shanghai, China
| |
Collapse
|
42
|
Kommineni N, Saka R, Bulbake U, Khan W. Cabazitaxel and thymoquinone co-loaded lipospheres as a synergistic combination for breast cancer. Chem Phys Lipids 2018; 224:104707. [PMID: 30521787 DOI: 10.1016/j.chemphyslip.2018.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 09/19/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Cabazitaxel as microtubule inhibitor and thymoquinone as HDAC inhibitor affects the important genes like p53, STAT3, Bax, BCL-2, p21 and down regulation of NF-κB are reported for potential activity against breast tumors. However, poor aqueous solubility and permeability hinders the delivery of these drugs to target site. To address the delivery challenges cabazitaxel and thymoquinone co-loaded lipospheres were developed. Lipospheres are the lipid based self-assemblies of particle size below 150 nm were prepared with more than 90% entrapment efficiency for both the drugs. In vitro drug release studies revealed there was a sustained diffusion controlled drug release from liposphere matrix leading to decrease in particle size with increase in zeta potential. Cytotoxicity studies on MCF-7 and MDA-MB-231 cells demonstrated cabazitaxel and thymoquinone as synergistic combination for the treatment of breast cancer which was proved by CompuSyn software. Enhanced efficacy of developed lipospheres can be due to rapid cellular internalization which was observed in confocal laser scanning microscopy. Drastic changes in cancer cell morphology such as nuclear fragmentation were observed upon treatment with these lipospheres in comparison to combination solution as observed in fluorescent imaging which are the hall marks of apoptosis. Cell cycle analysis and apoptosis studies confirmed the increased Sub G1 phase arrest as well as cell death due to apoptosis. Thus, as per observed results, it can be concluded that cabazitaxel and thymoquinone co-loaded lipospheres are the efficient delivery vehicles in management of breast cancer.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Raju Saka
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Upendra Bulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, 500037, India.
| |
Collapse
|
43
|
Fernandes C, Suares D, Yergeri MC. Tumor Microenvironment Targeted Nanotherapy. Front Pharmacol 2018; 9:1230. [PMID: 30429787 PMCID: PMC6220447 DOI: 10.3389/fphar.2018.01230] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Recent developments in nanotechnology have brought new approaches to cancer diagnosis and therapy. While enhanced permeability and retention effect promotes nano-chemotherapeutics extravasation, the abnormal tumor vasculature, high interstitial pressure and dense stroma structure limit homogeneous intratumoral distribution of nano-chemotherapeutics and compromise their imaging and therapeutic effect. Moreover, heterogeneous distribution of nano-chemotherapeutics in non-tumor-stroma cells damages the non-tumor cells, and interferes with tumor-stroma crosstalk. This can lead not only to inhibition of tumor progression, but can also paradoxically induce acquired resistance and facilitate tumor cell proliferation and metastasis. Overall, the tumor microenvironment plays a vital role in regulating nano-chemotherapeutics distribution and their biological effects. In this review, the barriers in tumor microenvironment, its consequential effects on nano-chemotherapeutics, considerations to improve nano-chemotherapeutics delivery and combinatory strategies to overcome acquired resistance induced by tumor microenvironment have been summarized. The various strategies viz., nanotechnology based approach as well as ligand-mediated, redox-responsive, and enzyme-mediated based combinatorial nanoapproaches have been discussed in this review.
Collapse
Affiliation(s)
| | | | - Mayur C Yergeri
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies - NMIMS, Mumbai, India
| |
Collapse
|
44
|
Kruger CA, Abrahamse H. Utilisation of Targeted Nanoparticle Photosensitiser Drug Delivery Systems for the Enhancement of Photodynamic Therapy. Molecules 2018; 23:E2628. [PMID: 30322132 PMCID: PMC6222717 DOI: 10.3390/molecules23102628] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/12/2018] [Accepted: 10/12/2018] [Indexed: 12/25/2022] Open
Abstract
The cancer incidence world-wide has caused an increase in the demand for effective forms of treatment. One unconventional form of treatment for cancer is photodynamic therapy (PDT). PDT has 3 fundamental factors, namely a photosensitiser (PS) drug, light and oxygen. When a PS drug is administered to a patient, it can either passively or actively accumulate within a tumour site and once exposed to a specific wavelength of light, it is excited to produce reactive oxygen species (ROS), resulting in tumour destruction. However, the efficacy of ROS generation for tumour damage is highly dependent on the uptake of the PS in tumour cells. Thus, PS selective/targeted uptake and delivery in tumour cells is a crucial factor in PDT cancer drug absorption studies. Generally, within non-targeted drug delivery mechanisms, only minor amounts of PS are able to passively accumulate in tumour sites (due to the enhanced permeability and retention (EPR) effect) and the remainder distributes into healthy tissues, causing unwanted side effects and poor treatment prognosis. Thus, to improve the efficacy of PDT cancer treatment, research is currently focused on the development of specific receptor-based PS-nanocarrier platform drugs, which promote the active uptake and absorption of PS drugs in tumour sites only, avoiding unwanted side effects, as well as treatment enhancement. Therefore, the aim of this review paper is to focus on current actively targeted or passively delivered PS nanoparticle drug delivery systems, that have been previously investigated for the PDT treatment of cancer and so to deduce their overall efficacy and recent advancements.
Collapse
Affiliation(s)
- Cherie Ann Kruger
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, Doornfontein 2001, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, Doornfontein 2001, South Africa.
| |
Collapse
|
45
|
Seshadri DR, Ramamurthi A. Nanotherapeutics to Modulate the Compromised Micro-Environment for Lung Cancers and Chronic Obstructive Pulmonary Disease. Front Pharmacol 2018; 9:759. [PMID: 30061830 PMCID: PMC6054931 DOI: 10.3389/fphar.2018.00759] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/22/2018] [Indexed: 11/13/2022] Open
Abstract
The use of nanomaterials to modulate the tumor microenvironment has great potential to advance outcomes in patients with lung cancer. Nanomaterials can be used to prolong the delivery time of therapeutics enabling their specific targeting to tumors while minimizing and potentially eliminating cytotoxic effects. Using nanomaterials to deliver small-molecule inhibitors for oncogene targeted therapy and cancer immunotherapy while concurrently enabling regeneration of the extracellular matrix could enhance our therapeutic reach and improve outcomes for patients with non-small cell lung cancer (NSCLC) and chronic obstructive pulmonary disease (COPD). The objective of this review is to highlight the role nanomedicines play in improving and reversing adverse outcomes in the tumor microenvironment for advancing treatments for targeting both diseases.
Collapse
Affiliation(s)
- Dhruv R. Seshadri
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Anand Ramamurthi
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, OH, United States
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
46
|
Alric C, Hervé-Aubert K, Aubrey N, Melouk S, Lajoie L, Même W, Même S, Courbebaisse Y, Ignatova AA, Feofanov AV, Chourpa I, Allard-Vannier E. Targeting HER2-breast tumors with scFv-decorated bimodal nanoprobes. J Nanobiotechnology 2018; 16:18. [PMID: 29466990 PMCID: PMC5820783 DOI: 10.1186/s12951-018-0341-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 02/09/2018] [Indexed: 01/22/2023] Open
Abstract
Background Recent advances in nanomedicine have shown the great interest of active targeting associated to nanoparticles. Single chain variable fragments (scFv) of disease-specific antibodies are very promising targeting entities because they are small, not immunogenic and able to bind their specific antigens. The present paper is devoted to biological properties in vitro and in vivo of fluorescent and pegylated iron oxide nanoparticles (SPIONs-Cy-PEG-scFv) functionalized with scFv targeting Human Epithelial growth Receptor 2 (HER2). Results Thanks to a site-selective scFv conjugation, the resultant nanoprobes demonstrated high affinity and specific binding to HER2 breast cancer cells. The cellular uptake of SPIONs-Cy-PEG-scFv was threefold higher than that for untargeted PEGylated iron oxide nanoparticles (SPIONs-Cy-PEG) and is correlated to the expression of HER2 on cells. In vivo, the decrease of MR signals in HER2+ xenograft tumor is about 30% at 24 h after the injection. Conclusions These results all indicate that SPIONs-Cy-PEG-scFv are relevant tumor-targeting magnetic resonance imaging agents, suitable for diagnosis of HER2 overexpressing breast tumor. Electronic supplementary material The online version of this article (10.1186/s12951-018-0341-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christophe Alric
- EA6295 'Nanomédicaments et Nanosondes', Université de Tours, 37200, Tours, France
| | - Katel Hervé-Aubert
- EA6295 'Nanomédicaments et Nanosondes', Université de Tours, 37200, Tours, France
| | - Nicolas Aubrey
- ISP, Université de Tours, INRA, UMR 1282, Equipe BIOMédicaments Anti-Parasitaires, 37380, Nouzilly, France
| | - Souad Melouk
- EA6295 'Nanomédicaments et Nanosondes', Université de Tours, 37200, Tours, France
| | - Laurie Lajoie
- GICC 'Groupe Innovation et Ciblage Cellulaire', Université de Tours, Equipe FRAME - Fc Récepteurs, Anticorps et MicroEnvironnement, 37032, Tours, France
| | - William Même
- CBM, CNRS, UPR4301, Equipe Complexes Métalliques et IRM pour applications biomédicales, 45071, Orléans, France
| | - Sandra Même
- CBM, CNRS, UPR4301, Equipe Complexes Métalliques et IRM pour applications biomédicales, 45071, Orléans, France
| | | | - Anastasia A Ignatova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya, 16/10, Moscow, 117997, Russia.,Biological Faculty, Lomonosov Moscow State University, Vorobyevi Gori 1, Moscow, 119992, Russia
| | - Alexey V Feofanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya, 16/10, Moscow, 117997, Russia.,Biological Faculty, Lomonosov Moscow State University, Vorobyevi Gori 1, Moscow, 119992, Russia
| | - Igor Chourpa
- EA6295 'Nanomédicaments et Nanosondes', Université de Tours, 37200, Tours, France
| | | |
Collapse
|
47
|
Gutiérrez-Lovera C, Vázquez-Ríos AJ, Guerra-Varela J, Sánchez L, de la Fuente M. The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines. Genes (Basel) 2017; 8:E349. [PMID: 29182542 PMCID: PMC5748667 DOI: 10.3390/genes8120349] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
In the last few decades, the field of nanomedicine applied to cancer has revolutionized cancer treatment: several nanoformulations have already reached the market and are routinely being used in the clinical practice. In the case of genetic nanomedicines, i.e., designed to deliver gene therapies to cancer cells for therapeutic purposes, advances have been less impressive. This is because of the many barriers that limit the access of the therapeutic nucleic acids to their target site, and the lack of models that would allow for an improvement in the understanding of how nanocarriers can be tailored to overcome them. Zebrafish has important advantages as a model species for the study of anticancer therapies, and have a lot to offer regarding the rational development of efficient delivery of genetic nanomedicines, and hence increasing the chances of their successful translation. This review aims to provide an overview of the recent advances in the development of genetic anticancer nanomedicines, and of the zebrafish models that stand as promising tools to shed light on their mechanisms of action and overall potential in oncology.
Collapse
Affiliation(s)
- C Gutiérrez-Lovera
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - A J Vázquez-Ríos
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - J Guerra-Varela
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Geneaqua S.L., Lugo 27002, Spain.
| | - L Sánchez
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
| | - M de la Fuente
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| |
Collapse
|
48
|
Wu D, Si M, Xue HY, Wong HL. Nanomedicine applications in the treatment of breast cancer: current state of the art. Int J Nanomedicine 2017; 12:5879-5892. [PMID: 28860754 PMCID: PMC5566389 DOI: 10.2147/ijn.s123437] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common malignant disease in women worldwide, but the current drug therapy is far from optimal as indicated by the high death rate of breast cancer patients. Nanomedicine is a promising alternative for breast cancer treatment. Nanomedicine products such as Doxil® and Abraxane® have already been extensively used for breast cancer adjuvant therapy with favorable clinical outcomes. However, these products were originally designed for generic anticancer purpose and not specifically for breast cancer treatment. With better understanding of the molecular biology of breast cancer, a number of novel promising nanotherapeutic strategies and devices have been developed in recent years. In this review, we will first give an overview of the current breast cancer treatment and the updated status of nanomedicine use in clinical setting, then discuss the latest important trends in designing breast cancer nanomedicine, including passive and active cancer cell targeting, breast cancer stem cell targeting, tumor microenvironment-based nanotherapy and combination nanotherapy of drug-resistant breast cancer. Researchers may get insight from these strategies to design and develop nanomedicine that is more tailored for breast cancer to achieve further improvements in cancer specificity, antitumorigenic effect, antimetastasis effect and drug resistance reversal effect.
Collapse
Affiliation(s)
- Di Wu
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Mengjie Si
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Hui-Yi Xue
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Ho-Lun Wong
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| |
Collapse
|