1
|
Kandell RM, Wu JR, Kwon EJ. Reprograming Clots for In Vivo Chemical Targeting in Traumatic Brain Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2301738. [PMID: 38780012 PMCID: PMC11293973 DOI: 10.1002/adma.202301738] [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: 02/23/2023] [Revised: 04/12/2024] [Indexed: 05/25/2024]
Abstract
Traumatic brain injury (TBI) is a critical public health concern, yet there are no therapeutics available to improve long-term outcomes. Drug delivery to TBI remains a challenge due to the blood-brain barrier and increased intracranial pressure. In this work, a chemical targeting approach to improve delivery of materials to the injured brain, is developed. It is hypothesized that the provisional fibrin matrix can be harnessed as an injury-specific scaffold that can be targeted by materials via click chemistry. To accomplish this, the brain clot is engineered in situ by delivering fibrinogen modified with strained cyclooctyne (SCO) moieties, which incorporated into the injury lesion and is retained there for days. Improved intra-injury capture and retention of diverse, clickable azide-materials including a small molecule azide-dye, 40 kDa azide-PEG nanomaterial, and a therapeutic azide-protein in multiple dosing regimens is subsequently observed. To demonstrate therapeutic translation of this approach, a reduction in reactive oxygen species levels in the injured brain after delivery of the antioxidant catalase, is achieved. Further, colocalization between azide and SCO-fibrinogen is specific to the brain over off-target organs. Taken together, a chemical targeting strategy leveraging endogenous clot formation is established which can be applied to improve therapeutic delivery after TBI.
Collapse
Affiliation(s)
- Rebecca M. Kandell
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Jason R. Wu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Ester J. Kwon
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Zhang Y, Lu Y, Li Y, Xu Y, Song W. Poly(Glutamic Acid)-Engineered Nanoplatforms for Enhanced Cancer Phototherapy. Curr Drug Deliv 2024; 21:326-338. [PMID: 36650626 DOI: 10.2174/1567201820666230116164511] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 01/19/2023]
Abstract
Phototherapies, including photothermal therapy and photodynamic therapy, have gained booming development over the past several decades for their attractive non-invasiveness nature, negligible adverse effects, minimal systemic toxicity, and high spatial selectivity. Phototherapy usually requires three components: light irradiation, photosensitizers, and molecular oxygen. Photosensitizers can convert light energy into heat or reactive oxygen species, which can be used in the tumor-killing process. The direct application of photosensitizers in tumor therapy is restricted by their poor water solubility, fast clearance, severe toxicity, and low cellular uptake. The encapsulation of photosensitizers into nanostructures is an attractive strategy to overcome these critical limitations. Poly(glutamic acid) (PGA) is a kind of poly(amino acid)s containing the repeating units of glutamic acid. PGA has superiority for cancer treatment because of its good biocompatibility, low immunogenicity, and modulated pH responsiveness. The hydrophilicity nature of PGA allows the physical entrapment of photosensitizers and anticancer drugs via the construction of amphiphilic polymers. Moreover, the pendent carboxyl groups of PGA enable chemical conjugation with therapeutic agents. In this mini-review, we highlight the stateof- the-art design and fabrication of PGA-based nanoplatforms for phototherapy. We also discuss the potential challenges and future perspectives of phototherapy, and clinical translation of PGA-based nanomedicines.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Yicong Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai-201318, P. R. China
| | - Wenliang Song
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai-200093, P. R. China
| |
Collapse
|
3
|
Fadhel MN, Appak Baskoy S, Wang Y, Hysi E, Kolios MC. Use of photoacoustic imaging for monitoring vascular disrupting cancer treatments. JOURNAL OF BIOPHOTONICS 2023; 16:e202000209. [PMID: 32888381 DOI: 10.1002/jbio.202000209] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 06/11/2023]
Abstract
Vascular disrupting agents disrupt tumor vessels, blocking the nutritional and oxygen supply tumors need to thrive. This is achieved by damaging the endothelium lining of blood vessels, resulting in red blood cells (RBCs) entering the tumor parenchyma. RBCs present in the extracellular matrix are exposed to external stressors resulting in biochemical and physiological changes. The detection of these changes can be used to monitor the efficacy of cancer treatments. Spectroscopic photoacoustic (PA) imaging is an ideal candidate for probing RBCs due to their high optical absorption relative to surrounding tissue. The goal of this work is to use PA imaging to monitor the efficacy of the vascular disrupting agent 5,6-Dimethylxanthenone-4-acetic acid (DMXAA) through quantitative analysis. Then, 4T1 breast cancer cells were injected subcutaneously into the left hind leg of eight BALB/c mice. After 10 days, half of the mice were treated with 15 mg/kg of DMXAA and the other half were injected with saline. All mice were imaged using the VevoLAZR X PA system before treatment, 24 and 72 hours after treatment. The imaging was done at six wavelengths and linear spectral unmixing was applied to the PA images to quantify three forms of hemoglobin (oxy, deoxy and met-hemoglobin). After imaging, tumors were histologically processed and H&E and TUNEL staining were used to detect the tissue damage induced by the DMXAA treatment. The total hemoglobin concentration remained unchanged after treatment for the saline treated mice. For DMXAA treated mice, a 10% increase of deoxyhemoglobin concentration was detected 24 hours after treatment and a 22.6% decrease in total hemoglobin concentration was observed by 72 hours. A decrease in the PA spectral slope parameters was measured 24 hours after treatment. This suggests that DMXAA induces vascular damage, causing red blood cells to extravasate. Furthermore, H&E staining of the tumor showed areas of bleeding with erythrocyte deposition. These observations are further supported by the increase in TUNEL staining in DMXAA treated tumors, revealing increased cell death due to vascular disruption. This study demonstrates the capability of PA imaging to monitor tumor vessel disruption by the vascular disrupting agent DMXAA.
Collapse
Affiliation(s)
- Muhannad N Fadhel
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada
- Department of Physics, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Sila Appak Baskoy
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada
- Department of Physics, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yanjie Wang
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada
- Department of Physics, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Eno Hysi
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada
- Department of Physics, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Ryerson University, Toronto, Ontario, Canada
- Institute for Biomedical Engineering, Science and Technology (iBEST), Toronto, Ontario, Canada
- Department of Physics, Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Vascular disrupting agent-induced amplification of tumor targeting and prodrug activation boosts anti-tumor efficacy. Sci China Chem 2022. [DOI: 10.1007/s11426-022-1347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
5
|
Zhang Y, Song W, Lu Y, Xu Y, Wang C, Yu DG, Kim I. Recent Advances in Poly(α- L-glutamic acid)-Based Nanomaterials for Drug Delivery. Biomolecules 2022; 12:636. [PMID: 35625562 PMCID: PMC9138577 DOI: 10.3390/biom12050636] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/16/2022] [Accepted: 04/23/2022] [Indexed: 02/06/2023] Open
Abstract
Poly(α-L-glutamic acid) (PGA) is a class of synthetic polypeptides composed of the monomeric unit α-L-glutamic acid. Owing to their biocompatibility, biodegradability, and non-immunogenicity, PGA-based nanomaterials have been elaborately designed for drug delivery systems. Relevant studies including the latest research results on PGA-based nanomaterials for drug delivery have been discussed in this work. The following related topics are summarized as: (1) a brief description of the synthetic strategies of PGAs; (2) an elaborated presentation of the evolving applications of PGA in the areas of drug delivery, including the rational design, precise fabrication, and biological evaluation; (3) a profound discussion on the further development of PGA-based nanomaterials in drug delivery. In summary, the unique structures and superior properties enables PGA-based nanomaterials to represent as an enormous potential in biomaterials-related drug delivery areas.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Wenliang Song
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Yiming Lu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Yixin Xu
- School of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China; (Y.Z.); (Y.L.); (Y.X.)
| | - Changping Wang
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Deng-Guang Yu
- School of Materials Science & Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China;
| | - Il Kim
- Department of Polymer Science and Engineering, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
6
|
Tao W, Zhao D, Li G, Li L, Li S, Ye H, Tian C, Lu Y, Li S, Sun Y, He Z, Sun J. Artificial tumor microenvironment regulated by first hemorrhage for enhanced tumor targeting and then occlusion for synergistic bioactivation of hypoxia-sensitive platesomes. Acta Pharm Sin B 2022; 12:1487-1499. [PMID: 35530142 PMCID: PMC9069311 DOI: 10.1016/j.apsb.2021.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/20/2021] [Accepted: 07/03/2021] [Indexed: 12/17/2022] Open
Abstract
The unique characteristics of the tumor microenvironment (TME) could be exploited to develop antitumor nanomedicine strategies. However, in many cases, the actual therapeutic effect is far from reaching our expectations due to the notable tumor heterogeneity. Given the amplified characteristics of TME regulated by vascular disrupting agents (VDAs), nanomedicines may achieve unexpected improved efficacy. Herein, we fabricate platelet membrane-fusogenic liposomes (PML/DP&PPa), namely “platesomes”, which actively load the hypoxia-activated pro-prodrug DMG-PR104A (DP) and physically encapsulate the photosensitizer pyropheophorbide a (PPa). Considering the different stages of tumor vascular collapse and shutdown induced by a VDA combretastatin-A4 phosphate (CA4P), PML/DP&PPa is injected 3 h after intraperitoneal administration of CA4P. First, CA4P-mediated tumor hemorrhage amplifies the enhanced permeation and retention (EPR) effect, and the platesome-biological targeting further promotes the tumor accumulation of PML/DP&PPa. Besides, CA4P-induced vascular occlusion inhibits oxygen supply, followed by photodynamic therapy-caused acute tumor hypoxia. This prolonged extreme hypoxia contributes to the complete activation of DP and then high inhibitory effect on tumor growth and metastasis. Thus, such a combining strategy of artificially-regulated TME and bio-inspired platesomes pronouncedly improves tumor drug delivery and boosts tumor hypoxia-selective activation, and provides a preferable solution to high-efficiency cancer therapy.
Collapse
Affiliation(s)
- Wenhui Tao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongyang Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingxiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Songhao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hao Ye
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chutong Tian
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yutong Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuying Li
- Department of Pharmaceutical and Engineering, College of Pharmaceutical and Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinghua Sun
- Department of Pharmaceutics, College of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Corresponding author. Tel./fax: +86 24 23986321.
| |
Collapse
|
7
|
Zhao J, Song W, Tang Z, Chen X. Macromolecular Effects in Medicinal Chemistry ※. ACTA CHIMICA SINICA 2022. [DOI: 10.6023/a21120602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
8
|
Dong S, Tang Y, He P, Ma S, Song W, Deng M, Tang Z. Hydrophobic modified poly(
l
‐glutamic acid) graft copolymer micelles with ultrahigh drug loading capacity for anticancer drug delivery. POLYM INT 2021. [DOI: 10.1002/pi.6342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Si Dong
- Department of Chemistry Northeast Normal University Changchun PR China
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun PR China
| | - Yue Tang
- Department of Chemistry Northeast Normal University Changchun PR China
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun PR China
| | - Pan He
- School of Materials Science and Engineering Changchun University of Science and Technology Changchun PR China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun PR China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun PR China
| | - Mingxiao Deng
- Department of Chemistry Northeast Normal University Changchun PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun PR China
| |
Collapse
|
9
|
Cell membrane-camouflaged liposomes for tumor cell-selective glycans engineering and imaging in vivo. Proc Natl Acad Sci U S A 2021; 118:2022769118. [PMID: 34301864 DOI: 10.1073/pnas.2022769118] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The dynamic change of cell-surface glycans is involved in diverse biological and pathological events such as oncogenesis and metastasis. Despite tremendous efforts, it remains a great challenge to selectively distinguish and label glycans of different cancer cells or cancer subtypes. Inspired by biomimetic cell membrane-coating technology, herein, we construct pH-responsive azidosugar liposomes camouflaged with natural cancer-cell membrane for tumor cell-selective glycan engineering. With cancer cell-membrane camouflage, the biomimetic liposomes can prevent protein corona formation and evade phagocytosis of macrophages, facilitating metabolic glycans labeling in vivo. More importantly, due to multiple membrane receptors, the biomimetic liposomes have prominent cell selectivity to homotypic cancer cells, showing higher glycan-labeling efficacy than a single-ligand targeting strategy. Further in vitro and in vivo experiments indicate that cancer cell membrane-camouflaged azidosugar liposomes not only realize cell-selective glycan imaging of different cancer cells and triple-negative breast cancer subtypes but also do well in labeling metastatic tumors. Meanwhile, the strategy is also applicable to the use of tumor tissue-derived cell membranes, which shows the prospect for individual diagnosis and treatment. This work may pave a way for efficient cancer cell-selective engineering and visualization of glycans in vivo.
Collapse
|
10
|
Liu M, Wu C, Ke L, Li Z, Wu YL. Emerging Biomaterials-Based Strategies for Inhibiting Vasculature Function in Cancer Therapy. SMALL METHODS 2021; 5:e2100347. [PMID: 34927997 DOI: 10.1002/smtd.202100347] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Indexed: 06/14/2023]
Abstract
The constant feeding of oxygen and nutrients through the blood vasculature has a vital role in maintaining tumor growth. Interestingly, recent endeavors have shown that nanotherapeutics with the strategy to block tumor blood vessels feeding nutrients and oxygen for starvation therapy can be helpful in cancer treatment. However, this field has not been detailed. Hence, this review will present an exhaustive summary of the existing biomaterial based strategies to disrupt tumor vascular function for effective cancer treatment, including hydrogel or nanogel-mediated local arterial embolism, thrombosis activator loaded nano-material-mediated vascular occlusion and anti-vascular drugs that block tumor vascular function, which may be beneficial to the design of anti-cancer nanomedicine by targeting the tumor vascular system.
Collapse
Affiliation(s)
- Minting Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
11
|
Zhang Y, He P, Zhang P, Yi X, Xiao C, Chen X. Polypeptides-Drug Conjugates for Anticancer Therapy. Adv Healthc Mater 2021; 10:e2001974. [PMID: 33929786 DOI: 10.1002/adhm.202001974] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/30/2021] [Indexed: 12/15/2022]
Abstract
Polypeptides are an important class of biodegradable polymers that have been widely used in drug delivery field. Owing to the controllable synthesis and robust side chain-functionalization ability, polypeptides have long been ideal candidates for conjugation with anticancer drugs. The chemical conjugation of anticancer drugs with polypeptides, termed polypeptides-drug conjugates, has demonstrated several advantages in improving pharmacokinetics, enhancing drug targeting, and controlling drug release, thereby leading to enhanced therapeutic outcomes with reduced side toxicities. This review focuses on the recent advances in the design and preparation of polypeptides-drug conjugates for enhanced anticancer therapy. Strategies for conjugation of different types of drugs, including small-molecule chemotherapeutic drugs, proteins, vascular disrupting agents, and gas molecules, onto polypeptides backbone are summarized. Finally, the challenges and future perspectives on the development of innovative polypeptides-drug conjugates for clinical cancer treatment are also presented.
Collapse
Affiliation(s)
- Yu Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Pan He
- School of Materials Science and Engineering Changchun University of Science and Technology Changchun 130022 P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials Jilin Biomedical Polymers Engineering Laboratory Changchun Institute of Applied Chemistry Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
12
|
Disrupting tumour vasculature and recruitment of aPDL1-loaded platelets control tumour metastasis. Nat Commun 2021; 12:2773. [PMID: 33986264 PMCID: PMC8119987 DOI: 10.1038/s41467-021-22674-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 03/18/2021] [Indexed: 12/17/2022] Open
Abstract
Although therapies of cancer are advancing, it remains challenging for therapeutics to reach the sites of metastasis, which accounts for majority of cancer associated death. In this study, we have developed a strategy that guides an anti-programmed cell death-ligand 1 (aPDL1) antibody to accumulate in metastatic lesions to promote anti-tumour immune responses. Briefly, we have developed a combination in which Vadimezan disrupts tumour blood vessels of tumour metastases and facilitates the recruitment and activation of adoptively transferred aPDL1-conjugated platelets. In situ activated platelets generate aPDL1-decorated platelet-derived microparticles (PMP) that diffuse within the tumour and elicit immune responses. The proposed combination increases 10-fold aPDL1 antibody accumulation in lung metastases as compared to the intravenous administration of the antibody and enhances the magnitude of immune responses leading to improved antitumour effects. Cancer metastasis is the leading cause of death in patients, here, the authors show disrupting tumor vasculature could recruit and activate anti-PD-L1 engineered platelet at metastatic tumor sites to block the PD-1/PD-L1 crosstalk and enhance the anticancer immunotherapy.
Collapse
|
13
|
Liu L, O’Kelly D, Schuetze R, Carlson G, Zhou H, Trawick ML, Pinney KG, Mason RP. Non-Invasive Evaluation of Acute Effects of Tubulin Binding Agents: A Review of Imaging Vascular Disruption in Tumors. Molecules 2021; 26:2551. [PMID: 33925707 PMCID: PMC8125421 DOI: 10.3390/molecules26092551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Tumor vasculature proliferates rapidly, generally lacks pericyte coverage, and is uniquely fragile making it an attractive therapeutic target. A subset of small-molecule tubulin binding agents cause disaggregation of the endothelial cytoskeleton leading to enhanced vascular permeability generating increased interstitial pressure. The resulting vascular collapse and ischemia cause downstream hypoxia, ultimately leading to cell death and necrosis. Thus, local damage generates massive amplification and tumor destruction. The tumor vasculature is readily accessed and potentially a common target irrespective of disease site in the body. Development of a therapeutic approach and particularly next generation agents benefits from effective non-invasive assays. Imaging technologies offer varying degrees of sophistication and ease of implementation. This review considers technological strengths and weaknesses with examples from our own laboratory. Methods reveal vascular extent and patency, as well as insights into tissue viability, proliferation and necrosis. Spatiotemporal resolution ranges from cellular microscopy to single slice tomography and full three-dimensional views of whole tumors and measurements can be sufficiently rapid to reveal acute changes or long-term outcomes. Since imaging is non-invasive, each tumor may serve as its own control making investigations particularly efficient and rigorous. The concept of tumor vascular disruption was proposed over 30 years ago and it remains an active area of research.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Devin O’Kelly
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Regan Schuetze
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Graham Carlson
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Heling Zhou
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Kevin G. Pinney
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76798, USA; (G.C.); (M.L.T.); (K.G.P.)
| | - Ralph P. Mason
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (L.L.); (D.O.); (R.S.); (H.Z.)
| |
Collapse
|
14
|
Zhao D, Huang X, Zhang Z, Ding J, Cui Y, Chen X. Engineered nanomedicines for tumor vasculature blockade therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1691. [PMID: 33480163 DOI: 10.1002/wnan.1691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022]
Abstract
Tumor vasculature blockade therapy (TVBT), including angiogenesis inhibition, vascular disruption, and vascular infarction, provides a promising treatment modality for solid tumors. However, low selectivity, drug resistance, and possible severe side effects have limited the clinical transformation of TVBT. Engineered nanoparticles offer potential solutions, including prolonged circulation time, targeted transportation, and controlled release of TVBT agents. Moreover, engineered nanomedicines provide a promising combination platform of TVBT with chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, ultrasound therapy, and gene therapy. In this article, we offer a comprehensive summary of the current progress of engineered nanomedicines for TVBT and also discuss current deficiencies and future directions for TVBT development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Xu Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China.,Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Yan Cui
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
15
|
Dodt K, Driessen MD, Lamer S, Schlosser A, Lühmann T, Meinel L. A Complete and Versatile Protocol: Decoration of Cell-Derived Matrices with Mass-Encoded Peptides for Multiplexed Protease Activity Detection. ACS Biomater Sci Eng 2020; 6:6598-6617. [PMID: 33320595 DOI: 10.1021/acsbiomaterials.0c01134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This article provides guidance toward a platform technology for monitoring enzyme activity within the extracellular matrix (ECM) assessed by quantifying reporters secreted into the cell culture supernatant and analyzed by tandem mass spectrometry. The reporters are enzymatically and covalently bound to the ECM by transglutaminases (TG) using the peptide sequence of human insulin-like growth factor I's (IGF-I) D-domain which is known to be bound to the ECM by transglutaminase. The IGF-I D-domain sequence is followed by a peptide sequence cleaved by the intended target protease. This protease-sensitive peptide sequence (PSS) is cleaved off the ECM and can be used to monitor target-enzyme activity by employing a downstream mass tag designed according to isobaric mass encoding strategies, i.e., the combination of isotopically labeled, heavy amino acids. Thereby, cleavage events are linked to the appearance of encoded mass tags, readily allowing multiplexing. This article presents the design and synthesis of these mass reporters. It further aims at detailing the search for peptide sequences responding to target proteases to facilitate future work on enzyme activity measurement for enzymatic activities of hitherto unknown enzymes. In conclusion, the goal of this article is to arm scientists interested in measurements of local enzymatic activities within the ECM with robust protocols and background knowledge.
Collapse
Affiliation(s)
- Katharina Dodt
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
| | - Marc D Driessen
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
| | - Stephanie Lamer
- Rudolf-Virchow-Center for Experimental Biomedicine, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Andreas Schlosser
- Rudolf-Virchow-Center for Experimental Biomedicine, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Tessa Lühmann
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
| | - Lorenz Meinel
- Institute of Pharmacy and Food Chemistry, University of Wuerzburg, Am Hubland, 97074, Wuerzburg, Germany
| |
Collapse
|
16
|
Yu H, Bao Y, Xu C, Chen L, Tang Z. Poly(L-Glutamic Acid)-Drug Conjugates for Chemo- and Photodynamic Combination Therapy. Macromol Biosci 2020; 21:e2000192. [PMID: 33043592 DOI: 10.1002/mabi.202000192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/26/2020] [Indexed: 11/09/2022]
Abstract
Despite the polymeric vascular disrupting agent (poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol)/combretastatin A4) nanoparticles can efficiently inhibit cancer growth, their further application is still a challenge owing to the tumor recurrence and metastasis after treatment. In this study, two poly(L -glutamic acid)-drug conjugates for chemo-and photodynamic combination therapy are fabricated. PLG-g-mPEG-CA4 nanoparticles are prepared by combretastatin A4 (CA4) and poly(L -glutamic acid)-graft-methoxy poly(ethylene glycol) (PLG-g-mPEG) using the Yamaguchi esterification reaction. PLG-g-mPEG-TPP (TPP: 5, 10, 15, 20-tetraphenylporphyrin) nanoparticles are constructed using PLG-g-mPEG and amine porphyrin through condensation reaction between carboxyl group of PLG-g-mPEG and amino group of porphyrin. The results showed that PLG-g-mPEG-CA4 nanoparticles have good antitumor ability. PLG-g-mPEG-TPP nanoparticles can produce singlet oxygen under the laser irradiation. Moreover, the combined therapy of PLG-g-mPEG-CA4 and PLG-g-mPEG-TPP nanoparticles has higher antitumor effect than the single chemotherapy or the single photodynamic therapy in vitro. The combination of CA4 nondrug and photodynamic therapy provides a new insight for enhancing the tumor therapeutic effect with vascular disrupting agents and other therapy.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China.,Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Yanli Bao
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Caina Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Li Chen
- Department of Chemistry, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, P. R. China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| |
Collapse
|
17
|
Mei D, Gong L, Zou Y, Yang D, Liu H, Liang Y, Sun N, Zhao L, Zhang Q, Lin Z. Platelet membrane-cloaked paclitaxel-nanocrystals augment postoperative chemotherapeutical efficacy. J Control Release 2020; 324:341-353. [DOI: 10.1016/j.jconrel.2020.05.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/06/2020] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
|
18
|
Hong S, Zheng DW, Zhang C, Huang QX, Cheng SX, Zhang XZ. Vascular disrupting agent induced aggregation of gold nanoparticles for photothermally enhanced tumor vascular disruption. SCIENCE ADVANCES 2020; 6:eabb0020. [PMID: 32548273 PMCID: PMC7274768 DOI: 10.1126/sciadv.abb0020] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/03/2020] [Indexed: 05/08/2023]
Abstract
Although vascular disrupting agents (VDAs) have been extensively implemented in current clinical tumor therapy, the notable adverse events caused by long-term dosing severely limit the therapeutic efficacy. To improve this therapy, we report a strategy for VDA-induced aggregation of gold nanoparticles to further destroy tumor vascular by photothermal effect. This strategy could effectively disrupt tumor vascular and cut off the nutrition supply after just one treatment. In the murine tumor model, this strategy results in notable tumor growth inhibition and gives rise to a 92.7% suppression of tumor growth. Besides, enhanced vascular damage could also prevent cancer cells from distant metastasis. Moreover, compared with clinical therapies, this strategy still exhibits preferable tumor suppression and metastasis inhibition ability. These results indicate that this strategy has great potential in tumor treatment and could effectively enhance tumor vascular damage and avoid the side effects caused by frequent administration.
Collapse
|
19
|
Yi X, Zhou H, Zhang Z, Xiong S, Yang K. X-rays-optimized delivery of radiolabeled albumin for cancer theranostics. Biomaterials 2020; 233:119764. [PMID: 31927252 DOI: 10.1016/j.biomaterials.2020.119764] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/25/2019] [Accepted: 01/04/2020] [Indexed: 12/18/2022]
Abstract
Exploiting the specific biological behaviors of the metabolizable nano-drugs assisted by X-rays exposure will be benefit for the optimization of radiotherapy-based combination therapy. Herein, Human serum albumin (HSA) nanoparticle, a familiar and metabolizable nanomaterial, is selected to investigate the changes of tumor accumulation and retention under X-rays exposure. Caveolin-1, an important protein which has positive correlation with cell uptake of nanomaterials, is expressed increasingly under X-rays exposure, resulting the enhanced cell uptake and prolonged tumor retention of HSA nanoparticles. After being labeled by radioactive iodine-125, HSA could be used for SPECT/CT imaging of mice. Moreover, it discovered that 125I-HSA nanoparticles possess much longer-time retention time in pre-irradiated tumor than that of controlled tumor. Using this strategy, the therapeutic efficiency of 131I-HSA injected mice after irradiating their tumors by X-rays is better than that of opposite sequence treated mice. In order to further improve the targeting ability of HSA, GNQEQVSPLTLLKXC peptide (A15) is conjugated to HSA nanoparticles for targeting the thrombosis in the tumor tissue triggered by X-rays exposure, realizing the high tumor accumulation of 131I-HSA assisted by X-rays exposure. Therefore, taking advantage of the increased expression of Caveolin-1 and the induced thrombosis under X-rays exposure, we optimized the delivery of radiolabeled HSA via enhancing the cell uptake and prolonging tumor retention of HSA for cancer combination therapy. Our work make contribution to guide the clinical albumin based combination therapy.
Collapse
Affiliation(s)
- Xuan Yi
- School of Pharmacy, Jiangsu Key Laboratory of Inflammation and Molecular Drug Targets, Nantong University, Nantong, Jiangsu, 226001, China.
| | - Hailin Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Zheng Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Saisai Xiong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
20
|
Wei Q, Shen N, Yu H, Wang Y, Tang Z, Chen X. FXIIIa substrate peptide decorated BLZ945 nanoparticles for specifically remodeling tumor immunity. Biomater Sci 2020; 8:5666-5676. [DOI: 10.1039/d0bm00713g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A synergistic antitumor strategy using CA4-NPs and coagulation-targeting BLZ945 nanoparticles was established based on tumor targeting and tumor immunity regulation.
Collapse
Affiliation(s)
- Qi Wei
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| | - Yue Wang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun 130022
- PR China
| |
Collapse
|
21
|
Jiang J, Shen N, Ci T, Tang Z, Gu Z, Li G, Chen X. Combretastatin A4 Nanodrug-Induced MMP9 Amplification Boosts Tumor-Selective Release of Doxorubicin Prodrug. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904278. [PMID: 31549774 DOI: 10.1002/adma.201904278] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/27/2019] [Indexed: 06/10/2023]
Abstract
Tumor-associated enzyme-activated prodrugs can potentially improve the selectivity of chemotherapeutics. However, the paucity of tumor-associated enzymes which are essential for prodrug activation usually limits the antitumor potency. A cooperative strategy that utilizes combretastatin A4 nanodrug (CA4-NPs) and matrix metalloproteinase 9 (MMP9)-activated doxorubicin prodrug (MMP9-DOX-NPs) is developed. CA4 is a typical vascular disrupting agent that can selectively disrupt immature tumor blood vessels and exacerbate the tumor hypoxia state. After treatment with CA4-NPs, MMP9 expression can be significantly enhanced by 5.6-fold in treated tumors, which further boosts tumor-selective active drug release of MMP9-DOX-NPs by 3.7-fold in an orthotopic 4T1 mammary adenocarcinoma mouse model. The sequential delivery of CA4-NPs and MMP9-DOX-NPs exhibits enhanced antitumor efficacy with reduced systemic toxicity compared with the noncooperative controls.
Collapse
Affiliation(s)
- Jian Jiang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Tianyuan Ci
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), Jonsson Comprehensive Cancer Center, Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, 90095, USA
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- California NanoSystems Institute (CNSI), Jonsson Comprehensive Cancer Center, Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA, 90095, USA
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
22
|
Yu S, Chen Z, Zeng X, Chen X, Gu Z. Advances in nanomedicine for cancer starvation therapy. Theranostics 2019; 9:8026-8047. [PMID: 31754379 PMCID: PMC6857045 DOI: 10.7150/thno.38261] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/25/2019] [Indexed: 12/24/2022] Open
Abstract
Abnormal cell metabolism with vigorous nutrition consumption is one of the major physiological characteristics of cancers. As such, the strategy of cancer starvation therapy through blocking the blood supply, depleting glucose/oxygen and other critical nutrients of tumors has been widely studied to be an attractive way for cancer treatment. However, several undesirable properties of these agents, such as low targeting efficacy, undesired systemic side effects, elevated tumor hypoxia, induced drug resistance, and increased tumor metastasis risk, limit their future applications. The recent development of starving-nanotherapeutics combined with other therapeutic methods displayed the promising potential for overcoming the above drawbacks. This review highlights the recent advances of nanotherapeutic-based cancer starvation therapy and discusses the challenges and future prospects of these anticancer strategies.
Collapse
Affiliation(s)
- Shuangjiang Yu
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China. E-mail:
| | - Zhaowei Chen
- Department of Bioengineering, Jonsson Comprehensive Cancer Center, California Nanosystems Institute (CNSI), and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Xuan Zeng
- Department of Bioengineering, Jonsson Comprehensive Cancer Center, California Nanosystems Institute (CNSI), and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China. E-mail:
| | - Zhen Gu
- Department of Bioengineering, Jonsson Comprehensive Cancer Center, California Nanosystems Institute (CNSI), and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
23
|
Dorababu A. Recent Advances in Nanoformulated Chemotherapeutic Drug Delivery (2015‐2019). ChemistrySelect 2019. [DOI: 10.1002/slct.201901064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Atukuri Dorababu
- Department of ChemistrySRMPP Govt. First Grade College, Huvinahadagali, Ballari (Dt), Karnataka India – 583219
| |
Collapse
|
24
|
Liang Y, Hao Y, Wu Y, Zhou Z, Li J, Sun X, Liu YN. Integrated Hydrogel Platform for Programmed Antitumor Therapy Based on Near Infrared-Triggered Hyperthermia and Vascular Disruption. ACS APPLIED MATERIALS & INTERFACES 2019; 11:21381-21390. [PMID: 31141335 DOI: 10.1021/acsami.9b05536] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Complete tumor regression is a great challenge faced by single therapy of near-infrared (NIR)-triggered hyperthermia or vascular disrupting agents. An injectable nanocomposite (NC) hydrogel is rationally designed for combined anticancer therapy based on NIR-triggered hyperthermia and vascular disruption. The NC hydrogel, codelivered with Prussian blue (PB) nanoparticles and combretastatin A4 (CA4), has good shear-thinning, self-recovery, and excellent photothermal properties. Because of the remarkable tumor-site retention and sustained release of CA4 (about 10% over 12 days), the NC hydrogel has a tumor suppression rate of 99.6%. The programmed combinational therapy conveys the concept of "attack + guard", where PB-based NIR irradiation imposes intensive attack on most of cancer cells, and CA4 serves as a guard against the tumor growth by cutting off the energy supply. Moreover, the biosafety and eco-friendliness of the hydrogel platform pave the way toward clinical applications.
Collapse
|
25
|
Zeng Q, Liu Y, Song Y, Feng B, Xu P, Shan B, Liao Z, Liu K, Zhong Y, Chen L, Su D. A UHPLC-MS/MS method coupled with simple and efficient alkaline hydrolysis for free and total determination of conjugate nanomedicine: Pharmacokinetic and biodistribution study of poly (l-glutamic acid)-graft-methoxy poly (ethylene glycol)/combretastatin A4. J Pharm Biomed Anal 2019; 169:215-224. [PMID: 30877933 DOI: 10.1016/j.jpba.2019.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 02/26/2019] [Accepted: 03/01/2019] [Indexed: 01/22/2023]
Abstract
Poly (l-glutamic acid)-Combretastatin A4 conjugate (PLG-CA4) is a novel nano-anticancer drug. For macromolecule conjugate nanomedicine, its pharmacology mechanism is closely related to the pharmacokinetic profiles in vivo. It is a great significance that evaluates this polymer drug combined by covalently bound via studying the pharmacokinetics and distribution characteristics. Therefore, it is urgent to develop a simple, accurate and practical analytical method for such conjugated polymers combined by covalently bound. In this study, a simple and complete alkali hydrolysis was designed and optimized for the total CA4 concentrations obtained from PLG-CA4. Ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS) method with multiple-reaction monitoring (MRM) mode and the internal standard (IS) were adopted to develop a sensitive and accurate method satisfied both free and total determination of PLG-CA4 in biosamples. The method was validated which showed good linearity over a wide concentration range (R2 > 0.99), and the intra- and inter-day assay variability was less than 15% for CA4. The mean extraction recoveries of CA4 from plasma were all more than 80.0%. Furthermore, the method was applied to the study of pharmacokinetics (PK) and tissue distribution of PLG-CA4 in tumor-bearing nude mice. PLG-CA4 significantly prolonged retention time and enhanced distribution of CA4 in tumor.
Collapse
Affiliation(s)
- Qiang Zeng
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Yali Liu
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Yonggui Song
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Bingwei Feng
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Pengfei Xu
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Baixi Shan
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Zhou Liao
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Kuangyi Liu
- SCIEX, Analytical Instrument Trading Co., Office Room 502, 5/F, Bldg 1, 518 North FU quan Road, IBP Changning District, Shanghai, 200335, PR China
| | - Youbao Zhong
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Lai Chen
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China
| | - Dan Su
- College of Pharmacy, Laboratory Animal Science and Technology Center, Jiangxi University of Traditional Chinese Medicine, 1688 Meiling Road, Nanchang, 330004, PR China.
| |
Collapse
|
26
|
Zhang MK, Li CX, Wang SB, Liu T, Song XL, Yang XQ, Feng J, Zhang XZ. Tumor Starvation Induced Spatiotemporal Control over Chemotherapy for Synergistic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1803602. [PMID: 30370690 DOI: 10.1002/smll.201803602] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/10/2018] [Indexed: 06/08/2023]
Abstract
By integrating the characteristics of each therapy modality and material chemistry, a multitherapy modality is put forward: tumor starvation triggered synergism with sensitized chemotherapy. Following starvation-induced amplification of pathological abnormalities in tumors, chemotherapy is arranged to be locally activated and accurately reinforced to perfect multitherapy synergism from spatial and temporal perspectives. To this end, glucose oxidase (GOD) and a hypoxic prodrug of tirapazamine (TPZ) are loaded in acidity-decomposable calcium carbonate (CaCO3 ) nanoparticles concurrently tethered by hyaluronic acid. This hybrid nanotherapeutic shows a strong tendency to accumulate in tumors postinjection due to the cooperation between passive and active targeting mechanisms. The GOD-driven oxidation reaction deprives tumors of glucose for starvation therapy and concomitantly induces tumorous abnormality amplifications including elevated acidity and exacerbated hypoxia. Programmatically, the acidity amplification causes CaCO3 decomposition, offering not only spatial control over the liberation of embedded TPZ just within tumors but also the temporal control over timely chemotherapy initiation to match the occurrence of hypoxia amplification and thus benefiting perfect synergism between starvation therapy and chemotherapy.
Collapse
Affiliation(s)
- Ming-Kang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Shi-Bo Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Tao Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Lin Song
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong, University of Science and Technology, Wuhan, 430074, China
| | - Xiao-Quan Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong, University of Science and Technology, Wuhan, 430074, China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
27
|
Poly (l-glutamic acid)-g-methoxy poly (ethylene glycol)-gemcitabine conjugate improves the anticancer efficacy of gemcitabine. Int J Pharm 2018; 550:79-88. [PMID: 30138704 DOI: 10.1016/j.ijpharm.2018.08.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 07/28/2018] [Accepted: 08/18/2018] [Indexed: 02/06/2023]
Abstract
Gemcitabine is widely used for anticancer therapy. However, its short blood circulation time and poor stability greatly impair its application. To solve this problem, we prepared a poly (l-glutamic acid)-g-methoxy poly (ethylene glycol)-gemcitabine conjugate (l-Gem) with a 14.3 wt% drug-loading content. l-Gem showed concentration- and time-dependent cytotoxicity towards 4T1, LLC, MIA PaCa-2 and A2780 in vitro. Pharmacokinetic and biodistribution studies indicated that l-Gem had remarkably enhanced blood stability, prolonged blood circulation time and greatly improved selective tumor distribution compared with free gemcitabine. The area under the concentration-time curve from zero to infinity [AUC(0-∞)] of l-Gem in plasma was 43-fold higher than that of free gemcitabine. The AUC(0-∞) of the inactive metabolite, 2'-deoxy-2',2'-difluorouridine in the l-Gem group was ∼20% of that observed in the free gemcitabine group. The drug tumor accumulation ratio in the l-Gem group relative to the free gemcitabine group was 9.9 at 36 h, while the tumor AUC ratio was 15.8. Testing on Balb/C mice bearing the 4T1 tumor further demonstrated that l-Gem had significantly higher anticancer efficacy than free gemcitabine in vivo. These findings indicated that l-Gem has great potential for cancer treatment.
Collapse
|
28
|
Zhou H, Lv S, Zhang D, Deng M, Zhang X, Tang Z, Chen X. A polypeptide based podophyllotoxin conjugate for the treatment of multi drug resistant breast cancer with enhanced efficiency and minimal toxicity. Acta Biomater 2018; 73:388-399. [PMID: 29694920 DOI: 10.1016/j.actbio.2018.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 02/07/2023]
Abstract
Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown significant activity against P-glycoprotein (P-gp) mediated multi drug resistant cancer cells. However, because of the poor aqueous solubility and high toxicity, PPT cannot be used in clinical cancer therapy. In order to enhance the efficiency and reduce side effect of PPT, a polypeptide based PPT conjugate PLG-g-mPEG-PPT was developed and used for the treatment of multi drug resistant breast cancer. The PLG-g-mPEG-PPT was prepared by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol) (PLG-g-mPEG) via ester bonds. The PPT conjugates self-assembled into nanoparticles with average sizes about 100 nm in aqueous solution. Western blotting assay showed that the PLG-g-mPEG-PPT could effectively inhibit the expression of P-gp in the multiple drug resistant MCF-7/ADR cells. In vitro cytotoxicity assay indicated that the resistance index (RI) values of PLG-g-mPEG-PPT on different drug-resistant cancer cell lines exhibited 57-270 folds reduction than of traditional microtubule inhibitor chemotherapeutic drug PTX or DTX. Hemolysis assay demonstrated that the conjugation greatly decreased the hemolytic activity of free PPT. Maximum tolerated dose (MTD) of PLG-g-mPEG-PPT increased greatly (13.3 folds) as compared to that of free PPT. In vivo study showed that the PLG-g-mPEG-PPT conjugate remarkably enhanced the antitumor efficacy against MCF-7/ADR xenograft tumors with a tumor suppression rate (TSR) of 82.5%, displayed significantly improved anticancer efficacy as compared to free PPT (TSR = 37.1%) with minimal toxicity when both of the two formulations were used in MTD. STATEMENT OF SIGNIFICANCE The development of multiple drug resistance (MDR) of cancer cells is the main cause of chemotherapy failure. The over-expression of P-glycoprotein (P-gp) has been recognized to be the most important cause of MDR in cancer. Podophyllotoxin (PPT) is a chemotherapeutic agent which has shown strong activity against P-gp mediated multidrug resistant cancer cells by simultaneously inhibiting the over-expression of P-gp and the growth of cancer cells. However, PPT can not be used in clinical cancer treatment due to its poor aqueous solubility and high toxicity. Herein, we developed a polypeptide based PPT conjugate PLG-g-mPEG-PPT by conjugating PPT to poly(l-glutamic acid)-g-methoxy poly(ethylene glycol). The PLG-g-mPEG-PPT shows significantly decreased hemolytic activity, greatly improved maximum tolerated dose and remarkably enhanced antitumor efficacy against MCF-7/ADR xenograft tumors as compared to free PPT.
Collapse
|
29
|
Tao Y, Zhu L, Zhao Y, Yi X, Zhu L, Ge F, Mou X, Chen L, Sun L, Yang K. Nano-graphene oxide-manganese dioxide nanocomposites for overcoming tumor hypoxia and enhancing cancer radioisotope therapy. NANOSCALE 2018; 10:5114-5123. [PMID: 29487939 DOI: 10.1039/c7nr08747k] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
While radiotherapy (RT) is commonly used in clinics for cancer treatment, the therapeutic efficiency is not satisfactory owing to the existence of the hypoxic tumor microenvironment which seriously affects the efficiency of RT. Herein, we design polyethylene glycol (PEG)-modified reduced nano-graphene oxide-manganese dioxide (rGO-MnO2-PEG) nanocomposites to trigger oxygen generation from H2O2 to reduce the tumor hypoxic microenvironments. We use the radioisotope, 131I labeled rGO-MnO2-PEG nanocomposites as therapeutic agents for in vivo tumor radioisotope therapy (RIT), achieving excellent tumor killing and further enhancing the therapeutic efficiency of RIT. More importantly, the dissolution of MnO2 under acidic conditions and the redox process during the catalytic pathway of H2O2 decomposition in the cellular microenvironment direct to the production of an enormous amount of Mn2+ which has been used as a contrast agent for magnetic resonance imaging (MRI). Our proposed work provides a strategy to trigger oxygen formation via an internal stimulus to enhance imaging-guided RIT efficiency.
Collapse
Affiliation(s)
- Yugui Tao
- College of Biological and Chemical Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Yu S, Zhang D, He C, Sun W, Cao R, Cui S, Deng M, Gu Z, Chen X. Injectable Thermosensitive Polypeptide-Based CDDP-Complexed Hydrogel for Improving Localized Antitumor Efficacy. Biomacromolecules 2017; 18:4341-4348. [PMID: 29141405 DOI: 10.1021/acs.biomac.7b01374] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Shuangjiang Yu
- Key
Laboratory of Polymer Ecomaterials, Changchun Institute of Applied
Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Dianliang Zhang
- Key
Laboratory of Polymer Ecomaterials, Changchun Institute of Applied
Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department
of Chemistry, Northeast Normal University, Changchun 130022, China
| | - Chaoliang He
- Key
Laboratory of Polymer Ecomaterials, Changchun Institute of Applied
Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Wujin Sun
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Rangjuan Cao
- Department
of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Shusen Cui
- Department
of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun 130033, China
| | - Mingxiao Deng
- Department
of Chemistry, Northeast Normal University, Changchun 130022, China
| | - Zhen Gu
- Joint
Department of Biomedical Engineering, University of North Carolina at Chapel Hill, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Xuesi Chen
- Key
Laboratory of Polymer Ecomaterials, Changchun Institute of Applied
Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
31
|
Song Z, Han Z, Lv S, Chen C, Chen L, Yin L, Cheng J. Synthetic polypeptides: from polymer design to supramolecular assembly and biomedical application. Chem Soc Rev 2017; 46:6570-6599. [PMID: 28944387 DOI: 10.1039/c7cs00460e] [Citation(s) in RCA: 255] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Synthetic polypeptides from the ring-opening polymerization of N-carboxyanhydrides (NCAs) are one of the most important biomaterials. The unique features of these synthetic polypeptides, including their chemical diversity of side chains and their ability to form secondary structures, enable their broad applications in the field of gene delivery, drug delivery, bio-imaging, tissue engineering, and antimicrobials. In this review article, we summarize the recent advances in the design of polypeptide-based supramolecular structures, including complexes with nucleic acids, micelles, vesicles, hybrid nanoparticles, and hydrogels. We also highlight the progress in the chemical design of functional polypeptides, which plays a crucial role to manipulate their assembly behaviours and optimize their biomedical performances. Finally, we conclude the review by discussing the future opportunities in this field, including further studies on the secondary structures and cost-effective synthesis of polypeptide materials.
Collapse
Affiliation(s)
- Ziyuan Song
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Liu T, Zhang D, Song W, Tang Z, Zhu J, Ma Z, Wang X, Chen X, Tong T. A poly(l-glutamic acid)-combretastatin A4 conjugate for solid tumor therapy: Markedly improved therapeutic efficiency through its low tissue penetration in solid tumor. Acta Biomater 2017; 53:179-189. [PMID: 28167300 DOI: 10.1016/j.actbio.2017.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/30/2017] [Accepted: 02/01/2017] [Indexed: 01/13/2023]
Abstract
UNLABELLED Combretastatin A4 (CA4) is a leading agent in vascular disrupting strategies for tumor therapy. Although many small-molecule prodrugs of CA4 have been developed to improve its solubility, the overall therapeutic efficiency is moderate. A key reason for this is the reversible effect that CA4 has on tubulin as well as its rapid clearance from plasma and tissues. In this study, we proposed a poly(l-glutamic acid)-CA4 conjugate (PLG-CA4) nanomedicine to fulfill the requirements for fully liberating the potential of CA4 on tumor therapy. Enhanced accumulation and retention of CA4 in tumor tissue, especially, high distribution and gradual release around tumor blood vessels resulted in prolonged vascular disruption and markedly enhanced therapeutic efficiency. We examined and compared the therapeutic effect of PLG-CA4 and commercial combretastatin-A4 phosphate (CA4P) in a murine colon C26 tumor. PLG-CA4 showed significantly prolonged retention in plasma and tumor tissue. Most importantly, the PLG-CA4 was mainly distributed around the tumor vessels because of its low tissue penetration in solid tumor. Pathology tests showed that PLG-CA4 treatment resulted in persistent vascular disruption and tumor damage 72h after a single injection, this in contrast to CA4P treatment, which showed quick relapse at an equal dose. Tumor suppression tests showed that PLG-CA4 treatment resulted in a tumor suppression rate of 74%, which indicates a significant advantage when compared to tumor suppression rate of the CA4P group, which was 24%. This is the first time that an advantage of the polymeric CA4 nanomedicine with low tissue penetration for solid tumor therapy has been shown. Thus, the results presented in this study provide a new idea for enhancing the tumor therapeutic effect of vascular disrupting agents. STATEMENT OF SIGNIFICANCE Nanomedicine usually has low tissue penetration in solid tumors, which limits the efficacy of nanomedicine in most cases. But herein, we demonstrate a nanosized vascular disruptive agent (VDA) PLG-CA4 has supper advantages over small molecular combretastatin-A4 phosphate (CA4P) because the PLG-CA4 was mainly distributed around the tumor vessels due to its low tissue penetration in solid tumor.
Collapse
Affiliation(s)
- Tianzhou Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Jiaming Zhu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Zhiming Ma
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xudong Wang
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Ti Tong
- Department of Thoracic Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
33
|
Sun Q, Zhou Z, Qiu N, Shen Y. Rational Design of Cancer Nanomedicine: Nanoproperty Integration and Synchronization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1606628. [PMID: 28234430 DOI: 10.1002/adma.201606628] [Citation(s) in RCA: 733] [Impact Index Per Article: 91.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Indexed: 05/21/2023]
Abstract
Current cancer nanomedicines can only mitigate adverse effects but fail to enhance therapeutic efficacies of anticancer drugs. Rational design of next-generation cancer nanomedicines should aim to enhance their therapeutic efficacies. Taking this into account, this review first analyzes the typical cancer-drug-delivery process of an intravenously administered nanomedicine and concludes that the delivery involves a five-step CAPIR cascade and that high efficiency at every step is critical to guarantee high overall therapeutic efficiency. Further analysis shows that the nanoproperties needed in each step for a nanomedicine to maximize its efficiency are different and even opposing in different steps, particularly what the authors call the PEG, surface-charge, size and stability dilemmas. To resolve those dilemmas in order to integrate all needed nanoproperties into one nanomedicine, stability, surface and size nanoproperty transitions (3S transitions for short) are proposed and the reported strategies to realize these transitions are comprehensively summarized. Examples of nanomedicines capable of the 3S transitions are discussed, as are future research directions to design high-performance cancer nanomedicines and their clinical translations.
Collapse
Affiliation(s)
- Qihang Sun
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Zhuxian Zhou
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Nasha Qiu
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Zheda Road 38, 310027, Hangzhou, China
| |
Collapse
|
34
|
Wang H, Wu J, Xie K, Fang T, Chen C, Xie H, Zhou L, Zheng S. Precise Engineering of Prodrug Cocktails into Single Polymeric Nanoparticles for Combination Cancer Therapy: Extended and Sequentially Controllable Drug Release. ACS APPLIED MATERIALS & INTERFACES 2017; 9:10567-10576. [PMID: 28271714 DOI: 10.1021/acsami.7b01938] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The synergistic combination of two or more chemotherapeutics frequently requires packaging in single delivery vehicles for the sequential release of each substance in a predictable manner. Here, we demonstrate for the first time that the rational engineering of a prodrug cocktail into single polymeric nanoparticles (NPs) can enable the sequential release of chemotherapeutics in a controllable manner. Exploiting combretastatin-A4 (CA4, 1) as a model antiangiogenesis agent, two ester derivatives, 2 and 3, tethered with saturated fatty acids (butanoic and heptanoic acid for 2 and 3, respectively) were synthesized. 7-Ethyl-10-hydroxycamptothecin (SN38) derivative 4, esterified with α-linolenic acid, was used as a cytotoxic drug. Because of their augmented lipophilicity and miscibility, all constructed prodrugs readily assembled with clinically approved polymeric matrices. Results showed that altering the aliphatic chains of modifiers for CA4 chemical derivatization enabled the drug retention capacity within particle systems to be adjusted, leading to the identification of the prodrug cocktail of 2 and 4 as an optimal combination for subsequent preclinical studies. Furthermore, in vivo assessements indicated that the resulting NPs simultaneously formulating 2 and 4 exhibited synergistic activities and outperformed NPs loaded with individual prodrugs 2 or 4 in terms of therapeutic efficacy. These findings highlight a novel and versatile strategy for tailoring chemically disparate prodrug cocktails for adaptation within a single nanoplatform as a potential modality for synergistic cancer therapy.
Collapse
Affiliation(s)
- Hangxiang Wang
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Jiaping Wu
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Ke Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Tao Fang
- Jinhua People's Hospital , Jinhua, Zhejiang Province 321000, P.R. China
| | - Chao Chen
- College of Life Sciences, Huzhou University , Huzhou, Zhejiang Province 313000, P.R. China
| | - Haiyang Xie
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Lin Zhou
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| | - Shusen Zheng
- The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health and Key Laboratory of Organ Transplantation of Zhejiang Province, School of Medicine, Zhejiang University , Hangzhou 310003, P.R. China
| |
Collapse
|
35
|
Yan J, Zhang D, Yu H, Ma L, Deng M, Tang Z, Zhang X. Patupilone-loaded poly(L-glutamic acid)-graft-methoxy-poly(ethylene glycol) micelle for oncotherapy. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:394-414. [DOI: 10.1080/09205063.2016.1277827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jing Yan
- Department of Chemistry, Xiangtan University, Xiangtan, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Dawei Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Mingxiao Deng
- College of Chemistry, Northeast Normal University, Changchun, PR China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, PR China
| | - Xuefei Zhang
- Department of Chemistry, Xiangtan University, Xiangtan, PR China
- Key Laboratory of Environmentally Friendly Chemistry and Applications of Ministry of Education, Key Laboratory of Polymeric Materials and Application Technology of Hunan Province, Xiangtan University, Xiangtan, PR China
| |
Collapse
|
36
|
Gârea SA, Voicu AI, Iovu H. Clay–Polymer Nanocomposites for Controlled Drug Release. CLAY-POLYMER NANOCOMPOSITES 2017:475-509. [DOI: 10.1016/b978-0-323-46153-5.00014-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
37
|
Rajan M, Krishnan P, Pradeepkumar P, Jeyanthinath M, Jeyaraj M, Ling MP, Arulselvan P, Higuchi A, Munusamy MA, Arumugam R, Benelli G, Murugan K, Kumar SS. Magneto-chemotherapy for cervical cancer treatment with camptothecin loaded Fe3O4 functionalized β-cyclodextrin nanovehicle. RSC Adv 2017. [DOI: 10.1039/c7ra06615e] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We portray a novel way to synthesis of iron oxide magnetic nanoparticle incorporated β-cyclodextrin (β-CD) nanocarrier stabilized by ethylenediamine tetra acetic acid (EDTA) obtaining remarkable biocompatibility and biodegradability.
Collapse
|
38
|
Chen J, Liang H, Lin L, Guo Z, Sun P, Chen M, Tian H, Deng M, Chen X. Gold-Nanorods-Based Gene Carriers with the Capability of Photoacoustic Imaging and Photothermal Therapy. ACS APPLIED MATERIALS & INTERFACES 2016; 8:31558-31566. [PMID: 27775317 DOI: 10.1021/acsami.6b10166] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Multifunctional nanoparticles with high gene transfection activity, low cytotoxicity, photoacoustic imaging ability, and photothermal therapeutic properties were prepared by conjugating low-molecular-weight polyethylenimine onto the surfaces of gold nanorods through the formation of stable S-Au bonded conjugates. Results revealed that the gene transfection efficiency of the prepared polyethylenimine-modified gold nanorods (GNRs-PEI1.8k) was higher and their cytotoxicity was less than those of the commercial reagent PEI25k. GNRs-PEI1.8k could also be potentially used as a photoacoustic and photothermal reagent to evaluate the pharmacokinetics, biodistribution, and antitumor effects of gene/drug nanoparticles. Therefore, GNRs-PEI1.8k can be considered a promising candidate for the clinical diagnosis and treatment of tumors.
Collapse
Affiliation(s)
- Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
| | - Hong Liang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
- Department of Chemistry, Northeast Normal University , Changchun 130024, China
| | - Lin Lin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
| | - Zhaopei Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao 999078, China
| | - Pingjie Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
| | - Meiwan Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao 999078, China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
| | - Mingxiao Deng
- Department of Chemistry, Northeast Normal University , Changchun 130024, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, China
| |
Collapse
|
39
|
|