1
|
Salah AN, Hashem AH, Zaki MB, Abulsoud AI, Atta AM, Elkalla WS, Moustafa HAM, El-Dakroury WA, El-Tokhy FS, ElBoghdady JA, Rizk NI, Abdel Mageed SS, Mohammed OA, Abdel-Reheim MA, Alghamdi HO, Doghish AS. Targeted Therapies: The Role of Monoclonal Antibodies in Disease Management. J Biochem Mol Toxicol 2025; 39:e70163. [PMID: 39887821 DOI: 10.1002/jbt.70163] [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: 12/08/2024] [Revised: 12/28/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Monoclonal antibodies (mAbs) are a key class of biotherapeutic medicines used to treat a wide range of diseases, such as cancer, infectious diseases, autoimmune disorders, cardiovascular diseases, and hemophilia. They can be engineered for greater effectiveness and specific applications while maintaining their structural elements for immune targeting. Traditional immunoglobulin treatments have limited therapeutic uses and various adverse effects. That makes mAbs show rapid growth in the pharmaceutical market, with over 250 mAbs in clinical studies. Although mAbs offer higher specificity, they are less effective against complex antigens. They have become essential in treating diseases with limited medical options, providing innovative solutions that improve patients' quality of life through increasing survival rates, shortening the length of stay in hospitals with an improved treatment outcome, and reducing side effects. This review outlines the mechanisms, applications, and advancements of mAbs, highlighting their transformative role in modern medicine and their potential to shape future therapeutic interventions.
Collapse
Affiliation(s)
- Akram N Salah
- Microbiology and Immunology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menofia, 32897, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Menoufia National University, km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Asmaa M Atta
- Pharmaceutical Chemistry Department, School of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Wagiha S Elkalla
- Microbiology and Immunology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Fatma Sa'eed El-Tokhy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Jasmine A ElBoghdady
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Huda O Alghamdi
- College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Egypt
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
2
|
Das S, Pattnaik G, Pattanaik S, Jena BR, Satapathy BS, Pradhan A. Envisioning Clinical Management of Breast Cancer: a Comprehensive Review. Curr Drug Discov Technol 2025; 22:e290424229495. [PMID: 38685777 DOI: 10.2174/0115701638300812240417055802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Background: Coming to the edge of disease manufacturing in the twenty-- first-century, breast cancer occupies a terrifying scenario in the globe, especially in adult women. Its curiosity endeavours remarkable advances made during the past decennaries for cancer treatment and diagnosis. OBJECTIVE It accounts for the fifth leading cause of transience, killing approximately 570,000 people per annum. To reduce the prognosis of clinical oncological development with the application of a new chemical entity, some of the critical challenges, like active pharmaceutical ingredients with high chemical resistance, extreme side effects, and high treatment costs are some of the limitations in the curbing aspects of breast melanoma. METHODS In cancer research, hence, the development of drugs that are safe, efficient, and cost-effective remains a 'Holy Grail' that may be considered as a boon to target the malignant tissues with novel therapeutics devices. RESULTS Through the findings on overcoming the drawbacks of traditional methods, researchers have given special attention to cancer-preventive and theranostic approaches based on some novel drug delivery systems. CONCLUSION The present study forecasts the wide-ranging modern applications, and on developing some novel liposomal drug delivery therapy against breast cancer.
Collapse
Affiliation(s)
- Shubhashree Das
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Gurudutta Pattnaik
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Sovan Pattanaik
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Jatani, 752050, Odisha, India
| | - Bikash Ranjan Jena
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Bhabani Sankar Satapathy
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Jatani, 752050, Odisha, India
| | - Ayushi Pradhan
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| |
Collapse
|
3
|
Zhang B, Wang W, Song Y, Chen H, Lin X, Chen J, Chen Y, Huang J, Li D, Wu S. Exploring the Mechanism of Sempervirine Inhibiting Glioblastoma Invasion Based on Network Pharmacology and Bioinformatics. Pharmaceuticals (Basel) 2024; 17:1318. [PMID: 39458959 PMCID: PMC11510114 DOI: 10.3390/ph17101318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/19/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Invasion is an important characteristic of the malignancy of glioblastoma (GBM) and a significant prognostic factor. Sempervirine (SPV), a yohimbine-type alkaloid, has been proven to inhibit GBM cells proliferation in previous research and found to have a potential effect in anti-invasion, but its mechanism of anti-invasion is still unknown. Methods: To explore its pharmacodynamics in inhibiting GBM cell invasion in this study, we combined network pharmacology and bioinformatics to comprehensive exploratory analysis of SPV and verified the mechanism in vitro. Results: Firstly, targets of SPV and invasion-related genes were collected from public databases. Moreover, GBM samples were obtained to analyze differentially expressed genes (DEGs) from The Cancer Genome Atlas (TCGA). Then, the relevant targets of SPV inhibiting GBM invasion (SIGI) were obtained through the intersection of the three gene sets. Further, GO and KEGG analysis showed that the targets of SIGI were heavily enriched in the AKT signaling pathway. Subsequently, based on the method of machine learning, a clinical prognostic model of the relevant targets of SIGI was constructed using GBM samples from TCGA and the Gene Expression Omnibus (GEO). A four-genes model (DUSP6, BMP2, MMP2, and MMP13) was successfully constructed, and Vina Scores of MMP2 and MMP13 in molecular docking were higher, which may be the main targets of SIGI. Then, the effect of SIGI was confirmed via functional experiments on invasion, migration, and adhesion assay, and the effect involved changes in the expressions of p-AKT, MMP2 and MMP13. Finally, combined with AKT activator (SC79) and inhibitor (MK2206), we further confirmed that SPV inhibits GBM invasion through AKT phosphorylation. Conclusions: This study provides valuable and an expected point of view into the regulation of AKT phosphorylation and inhibition of GBM invasion by SPV.
Collapse
Affiliation(s)
- Bingqiang Zhang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Wenyi Wang
- Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Yu Song
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Huixian Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Xinxin Lin
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Jingjing Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Ying Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Jinfang Huang
- Fuzhou First General Hospital, Fuzhou 350009, China;
| | - Desen Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| | - Shuisheng Wu
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China; (B.Z.); (Y.S.); (H.C.); (X.L.); (J.C.); (Y.C.)
| |
Collapse
|
4
|
Liu Z, Ma K, Jia Q, Yang Y, Fan P, Wang Y, Wang J, Sun J, Sun L, Shi H, Sun L, Zhu B, Xu W, Zhang L, Jain RK, Qin S, Huang Y. Baseline tumour vessel perfusion as a non-invasive predictive biomarker for immune checkpoint therapy in non-small-cell lung cancer. BMJ ONCOLOGY 2024; 3:e000473. [PMID: 39886162 PMCID: PMC11347692 DOI: 10.1136/bmjonc-2024-000473] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 02/01/2025]
Abstract
Objective Current biomarkers for predicting immunotherapy response in non-small-cell lung cancer (NSCLC) are derived from invasive procedures with limited predictive accuracy. Thus, identifying a non-invasive predictive biomarker would improve patient stratification and precision immunotherapy. Methods and analysis In this retrospective multicohort study, the discovery cohort included 205 NSCLC patients screened from ORIENT-11 and an external validation (EV) cohort included 99 real-world NSCLC patients. The 'onion-mode segmentation' method was developed to extract 'onion-mode perfusion' (OMP) from contrast-enhanced CT images. The predictive performance of OMP or its combination with the PD-L1 Tumour Proportion Score (TPS) was evaluated by the area under the curve (AUC). Results High baseline OMP was associated with significantly longer survival and predicted patient response to combination anti-PD-(L)1 therapy in the discovery and EV cohorts. OMP complemented the PD-L1 TPS with superior predictive sensitivity (p=0.02). In the PD-L1 TPS<50% subgroup, OMP achieved an AUC of 0.77 for the estimation of treatment response (95% CI 0.66 to 0.86, p<0.0001). A simple bivariate model of OMP/PD-L1 robustly predicted therapeutic response in both the discovery (AUC 0.82, 95% CI 0.74 to 0.88, p<0.0001) and EV (AUC 0.80, 95% CI 0.67 to 0.89, p<0.0001) cohorts. Conclusion OMP, derived from routine CT examination, could serve as a non-invasive and cost-effective biomarker to predict NSCLC patient response to immune checkpoint inhibitor-based therapy. OMP could be used alone or in combination with other biomarkers to improve precision immunotherapy.
Collapse
Affiliation(s)
- Zhenhua Liu
- Department of Radiotherapy, State Key Laboratory of Radiation Medicine and Prevention, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy, Yancheng First Hospital Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu, China
| | - Ke Ma
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Qingzhu Jia
- Institute of Cancer, Third Military Medical University, Chongqing, Chongqing, China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Peng Fan
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Ying Wang
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Junhui Wang
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Jiya Sun
- New Drug Biology and Translational Medicine, Innovent Biologics Inc, Suzhou, Jiangsu, China
| | - Liansai Sun
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Hongtai Shi
- Department of Radiation Oncology, Yancheng Third People’s Hospital, Yancheng, Jiangsu, China
| | - Liang Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu, China
| | - Bo Zhu
- Institute of Cancer, Third Military Medical University, Chongqing, Chongqing, China
| | - Wei Xu
- New Drug Biology and Translational Medicine, Innovent Biologics Inc, Suzhou, Jiangsu, China
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Rakesh K. Jain
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Songbing Qin
- Department of Radiotherapy, State Key Laboratory of Radiation Medicine and Prevention, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yuhui Huang
- Cyrus Tang Medical Institute, State Key Laboratory of Radiation Medicine and Prevention, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
5
|
Yang Q, Zhou X, Lou B, Zheng N, Chen J, Yang G. An F OF 1-ATPase motor-embedded chromatophore as a nanorobot for overcoming biological barriers and targeting acidic tumor sites. Acta Biomater 2024; 179:207-219. [PMID: 38513724 DOI: 10.1016/j.actbio.2024.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024]
Abstract
Despite the booming progress of anticancer nanomedicines in the past two decades, precise tumor-targetability and sufficient tumor-accumulation are less successful and still require further research. To tackle this challenge, herein we present a biomolecular motor (FOF1-ATPase)-embedded chromatophore as nanorobot to efficiently overcome biological barriers, and thoroughly investigate its chemotactic motility, tumor-accumulation ability and endocytosis. Chromatophores embedded with FOF1-ATPase motors were firstly extracted from Thermus thermophilus, then their properties were fully characterized. Specifically, two microfluidic platforms (laminar flow microchip and tumor microenvironment (TME) microchip) were designed and developed to fully investigate the motility, tumor-accumulation ability and endocytosis of the chromatophore nanorobot (CN). The results from the laminar flow microchip indicated that the obtained CN possessed the strongly positive chemotaxis towards protons. And the TME microchip experiments verified that the CN had a desirable tumor-accumulation ability. Cellular uptake experiments demonstrated that the CN efficiently promoted the endocytosis of the fluorescence DiO into the HT-29 cells. And the in vivo studies revealed that the intravenously administered CN exhibited vigorous tumor-targetability and accumulation ability as well as highly efficient antitumor efficacy. All the results suggested that FOF1-ATPase motors-embedded CN could be promising nanomachines with powerful self-propulsion for overcoming physiological barriers and tumor-targeted drug delivery. STATEMENT OF SIGNIFICANCE: In this study, we demonstrated that FOF1-ATPase-embedded chromatophore nanorobots exhibit a strong proton chemotaxis, which not only plays a key role in tumor-targetability and accumulation, but also promotes tumor tissue penetration and internalization. The results of in vitro and in vivo studies indicated that drug-loaded chromatophore nanorobots are capable to simultaneously accomplish tumor-targeting, accumulation, penetration and internalization for enhanced tumor therapy. Our study provides a fundamental basis for further study on FOF1-ATPase-embedded chromatophore as tumor-targeting drug delivery systems that have promising clinical applications. It offers a new and more efficient delivery vehicle for cancer related therapeutics.
Collapse
Affiliation(s)
- Qingliang Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xuhui Zhou
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Bang Lou
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Ning Zheng
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Jiale Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
6
|
Dogra P, Shinglot V, Ruiz-Ramírez J, Cave J, Butner JD, Schiavone C, Duda DG, Kaseb AO, Chung C, Koay EJ, Cristini V, Ozpolat B, Calin GA, Wang Z. Translational modeling-based evidence for enhanced efficacy of standard-of-care drugs in combination with anti-microRNA-155 in non-small-cell lung cancer. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.14.24304306. [PMID: 38559070 PMCID: PMC10980136 DOI: 10.1101/2024.03.14.24304306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Elevated microRNA-155 (miR-155) expression in non-small-cell lung cancer (NSCLC) promotes cisplatin resistance and negatively impacts treatment outcomes. However, miR-155 can also boost anti-tumor immunity by suppressing PD-L1 expression. We developed a multiscale mechanistic model, calibrated with in vivo data and then extrapolated to humans, to investigate the therapeutic effects of nanoparticle-delivered anti-miR-155 in NSCLC, alone or in combination with standard-of-care drugs. Model simulations and analyses of the clinical scenario revealed that monotherapy with anti-miR-155 at a dose of 2.5 mg/kg administered once every three weeks has substantial anti-cancer activity. It led to a median progression-free survival (PFS) of 6.7 months, which compared favorably to cisplatin and immune checkpoint inhibitors. Further, we explored the combinations of anti-miR-155 with standard-of-care drugs, and found strongly synergistic two- and three-drug combinations. A three-drug combination of anti-miR-155, cisplatin, and pembrolizumab resulted in a median PFS of 13.1 months, while a two-drug combination of anti-miR-155 and cisplatin resulted in a median PFS of 11.3 months, which emerged as a more practical option due to its simple design and cost-effectiveness. Our analyses also provided valuable insights into unfavorable dose ratios for drug combinations, highlighting the need for optimizing dose regimen to prevent antagonistic effects. Thus, this work bridges the gap between preclinical development and clinical translation of anti-miR-155 and unravels the potential of anti-miR-155 combination therapies in NSCLC.
Collapse
Affiliation(s)
- Prashant Dogra
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
| | - Vrushaly Shinglot
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Joseph Cave
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Joseph D. Butner
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmine Schiavone
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Dan G. Duda
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ahmed O. Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
- Department of Imaging Physics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - George A. Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Department of Medicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, USA
| |
Collapse
|
7
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
8
|
Delgado M, Garcia-Sanz JA. Therapeutic Monoclonal Antibodies against Cancer: Present and Future. Cells 2023; 12:2837. [PMID: 38132155 PMCID: PMC10741644 DOI: 10.3390/cells12242837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
A series of monoclonal antibodies with therapeutic potential against cancer have been generated and developed. Ninety-one are currently used in the clinics, either alone or in combination with chemotherapeutic agents or other antibodies, including immune checkpoint antibodies. These advances helped to coin the term personalized medicine or precision medicine. However, it seems evident that in addition to the current work on the analysis of mechanisms to overcome drug resistance, the use of different classes of antibodies (IgA, IgE, or IgM) instead of IgG, the engineering of the Ig molecules to increase their half-life, the acquisition of additional effector functions, or the advantages associated with the use of agonistic antibodies, to allow a broad prospective usage of precision medicine successfully, a strategy change is required. Here, we discuss our view on how these strategic changes should be implemented and consider their pros and cons using therapeutic antibodies against cancer as a model. The same strategy can be applied to therapeutic antibodies against other diseases, such as infectious or autoimmune diseases.
Collapse
Affiliation(s)
| | - Jose A. Garcia-Sanz
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), 28040 Madrid, Spain;
| |
Collapse
|
9
|
Davodabadi F, Mirinejad S, Fathi-Karkan S, Majidpour M, Ajalli N, Sheervalilou R, Sargazi S, Rozmus D, Rahdar A, Diez-Pascual AM. Aptamer-functionalized quantum dots as theranostic nanotools against cancer and bacterial infections: A comprehensive overview of recent trends. Biotechnol Prog 2023; 39:e3366. [PMID: 37222166 DOI: 10.1002/btpr.3366] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/21/2023] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
Aptamers (Apts) are synthetic nucleic acid ligands that can be engineered to target various molecules, including amino acids, proteins, and pharmaceuticals. Through a series of adsorption, recovery, and amplification steps, Apts are extracted from combinatorial libraries of synthesized nucleic acids. Using aptasensors in bioanalysis and biomedicine can be improved by combining them with nanomaterials. Moreover, Apt-associated nanomaterials, including liposomes, polymeric, dendrimers, carbon nanomaterials, silica, nanorods, magnetic NPs, and quantum dots (QDs), have been widely used as promising nanotools in biomedicine. Following surface modifications and conjugation with appropriate functional groups, these nanomaterials can be successfully used in aptasensing. Advanced biological assays can use Apts immobilized on QD surfaces through physical interaction and chemical bonding. Accordingly, modern QD aptasensing platforms rely on interactions between QDs, Apts, and targets to detect them. QD-Apt conjugates can be used to directly detect prostate, ovarian, colorectal, and lung cancers or simultaneously detect biomarkers associated with these malignancies. Tenascin-C, mucin 1, prostate-specific antigen, prostate-specific membrane antigen, nucleolin, growth factors, and exosomes are among the cancer biomarkers that can be sensitively detected using such bioconjugates. Furthermore, Apt-conjugated QDs have shown great potential for controlling bacterial infections such as Bacillus thuringiensis, Pseudomonas aeruginosa, Escherichia coli, Acinetobacter baumannii, Campylobacter jejuni, Staphylococcus aureus, and Salmonella typhimurium. This comprehensive review discusses recent advancements in the design of QD-Apt bioconjugates and their applications in cancer and bacterial theranostics.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sonia Fathi-Karkan
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mahdi Majidpour
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Narges Ajalli
- Department of Chemical Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | | | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Dominika Rozmus
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury, Olsztyn, Poland
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| | - Ana M Diez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Quimica Analitica, Quimica Fisica e Ingenieria Quimica, Madrid, Spain
| |
Collapse
|
10
|
Maddiboyina B, Ramaiah, Nakkala RK, Roy H. Perspectives on cutting-edge nanoparticulate drug delivery technologies based on lipids and their applications. Chem Biol Drug Des 2023; 102:377-394. [PMID: 36916008 DOI: 10.1111/cbdd.14230] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/27/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023]
Abstract
Numerous nanotech arenas in therapeutic biology have recently provided a scientific platform to manufacture a considerable swath of unique chemical entities focusing on drugs. Recently, nanoparticulate drug delivery systems have emerged to deliver a specific drug to a specified site. Among all other carriers, lipids possess features exclusive to nanostructured dosage forms. The bioavailability of orally administered drugs is typically negatively affected by their poor water solubility, resulting from the unique chemical moieties introduced. Because of their unique advantages, lipid nanoparticles must become increasingly predictable as a robust delivery mechanism. The enhanced biopharmaceutical properties and significance of lipid-based targeting technologies such as liposomes, niosomes, solid lipid nanoparticles and micelles are highlighted in this review. Pharmaceutical implications of lipid nanocarriers for the transport and distribution of various therapeutic agents, such as biotechnological products and small pharmaceutical molecules, is a booming topic. Lipid nanoparticles as drug delivery systems have many appealing properties, including high biocompatibility, ease of preparation, tissue specificity, avoidance of reticuloendothelial systems, delayed drug release, scale-up feasibility, nontoxicity and targeted delivery. The use of lipid nanoparticles to enhance the transport of biopharmaceuticals is currently considered state-of-the-art. Similarly, we critically examine the upcoming guidelines that therapeutic scientists should handle.
Collapse
Affiliation(s)
| | - Ramaiah
- Freyr Solutions, Phoenix SEZ, Hyderabad, Telangana, India
| | | | | |
Collapse
|
11
|
Wasson EM, He W, Ahlquist J, Hynes WF, Triplett MG, Hinckley A, Karelehto E, Gray-Sherr DR, Friedman CF, Robertson C, Shusteff M, Warren R, Coleman MA, Moya ML, Wheeler EK. A perfused multi-well bioreactor platform to assess tumor organoid response to a chemotherapeutic gradient. Front Bioeng Biotechnol 2023; 11:1193430. [PMID: 37324446 PMCID: PMC10264793 DOI: 10.3389/fbioe.2023.1193430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
There is an urgent need to develop new therapies for colorectal cancer that has metastasized to the liver and, more fundamentally, to develop improved preclinical platforms of colorectal cancer liver metastases (CRCLM) to screen therapies for efficacy. To this end, we developed a multi-well perfusable bioreactor capable of monitoring CRCLM patient-derived organoid response to a chemotherapeutic gradient. CRCLM patient-derived organoids were cultured in the multi-well bioreactor for 7 days and the subsequently established gradient in 5-fluorouracil (5-FU) concentration resulted in a lower IC50 in the region near the perfusion channel versus the region far from the channel. We compared behaviour of organoids in this platform to two commonly used PDO culture models: organoids in media and organoids in a static (no perfusion) hydrogel. The bioreactor IC50 values were significantly higher than IC50 values for organoids cultured in media whereas only the IC50 for organoids far from the channel were significantly different than organoids cultured in the static hydrogel condition. Using finite element simulations, we showed that the total dose delivered, calculated using area under the curve (AUC) was similar between platforms, however normalized viability was lower for the organoid in media condition than in the static gel and bioreactor. Our results highlight the utility of our multi-well bioreactor for studying organoid response to chemical gradients and demonstrate that comparing drug response across these different platforms is nontrivial.
Collapse
Affiliation(s)
- Elisa Marie Wasson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Wei He
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Jesse Ahlquist
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - William Fredrick Hynes
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Michael Gregory Triplett
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Aubree Hinckley
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Eveliina Karelehto
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | | | - Caleb Fisher Friedman
- Department of Computational Media, University of California Santa Cruz, Santa Cruz, CA, United States
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
- UC Davis Comprehensive Cancer Center, Davis, CA, United States
| | - Maxim Shusteff
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Robert Warren
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Matthew A. Coleman
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Monica Lizet Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| | - Elizabeth K. Wheeler
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States
| |
Collapse
|
12
|
Yin S, Lu R, Li Y, Sun D, Liu C, Liu B, Li J. A microfluidic device inspired by leaky tumor vessels for hematogenous metastasis mechanism research. Analyst 2023; 148:1570-1578. [PMID: 36892183 DOI: 10.1039/d2an02081e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Endothelial intercellular pores of tumor vessels generally lead to enhanced interstitial flow and may facilitate the migration of tumor cells. The permeability of tumor vessels causes a concentration gradient of growth factors (CGGF) from blood vessels to tumor tissues, which is opposite to the direction of interstitial flow. In this work, exogenous chemotaxis under the CGGF is demonstrated as a mechanism of hematogenous metastasis. A bionic microfluidic device inspired by endothelial intercellular pores of tumor vessels has been designed to study the mechanism. A porous membrane vertically integrated into the device using a novel compound mold is utilized to mimic the leaky vascular wall. The formation mechanism of the CGGF caused by endothelial intercellular pores is numerically analyzed and experimentally verified. The migration behavior of U-2OS cells is studied in the microfluidic device. The device is divided into three regions of interest (ROI): primary site, migration zone, and tumor vessel. The number of cells in the migration zone increases significantly under the CGGF, but decreases under no CGGF, indicating tumor cells may be guided to the vascellum by exogenous chemotaxis. Transendothelial migration is subsequently monitored, demonstrating the successful replication of the key steps in vitro in the metastatic cascade by the bionic microfluidic device.
Collapse
Affiliation(s)
- Shuqing Yin
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Ruoyu Lu
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Yang Li
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Dexian Sun
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| | - Chong Liu
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China. .,Key Laboratory for Precision and Non-traditional Machining Technology of Ministry of Education, Dalian University of Technology, Dalian, China
| | - Bo Liu
- School of Biomedical Engineering, Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China.
| | - Jingmin Li
- Key Laboratory for Micro/Nano Technology and System of Liaoning Province, Dalian University of Technology, Dalian, China.
| |
Collapse
|
13
|
Gileva A, Trushina D, Yagolovich A, Gasparian M, Kurbanova L, Smirnov I, Burov S, Markvicheva E. Doxorubicin-Loaded Polyelectrolyte Multilayer Capsules Modified with Antitumor DR5-Specific TRAIL Variant for Targeted Drug Delivery to Tumor Cells. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:902. [PMID: 36903780 PMCID: PMC10005140 DOI: 10.3390/nano13050902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 06/18/2023]
Abstract
Recently, biodegradable polyelectrolyte multilayer capsules (PMC) have been proposed for anticancer drug delivery. In many cases, microencapsulation allows to concentrate the substance locally and prolong its flow to the cells. To reduce systemic toxicity when delivering highly toxic drugs, such as doxorubicin (DOX), the development of a combined delivery system is of paramount importance. Many efforts have been made to exploit the DR5-dependent apoptosis induction for cancer treatment. However, despite having a high antitumor efficacy of the targeted tumor-specific DR5-B ligand, a DR5-specific TRAIL variant, its fast elimination from a body limits its potential use in a clinic. A combination of an antitumor effect of the DR5-B protein with DOX loaded in the capsules could allow to design a novel targeted drug delivery system. The aim of the study was to fabricate PMC loaded with a subtoxic concentration of DOX and functionalized with the DR5-B ligand and to evaluate a combined antitumor effect of this targeted drug delivery system in vitro. In this study, the effects of PMC surface modification with the DR5-B ligand on cell uptake both in 2D (monolayer culture) and 3D (tumor spheroids) were studied by confocal microscopy, flow cytometry and fluorimetry. Cytotoxicity of the capsules was evaluated using an MTT test. The capsules loaded with DOX and modified with DR5-B demonstrated synergistically enhanced cytotoxicity in both in vitro models. Thus, the use of the DR5-B-modified capsules loaded with DOX at a subtoxic concentration could provide both targeted drug delivery and a synergistic antitumor effect.
Collapse
Affiliation(s)
- Anastasia Gileva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Daria Trushina
- Laboratory of Bioorganic Structures, Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, 119333 Moscow, Russia
| | - Anne Yagolovich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - Marine Gasparian
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Leyli Kurbanova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Ivan Smirnov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Sergey Burov
- Cytomed JSC, Orlovo-Denisovsky pr. 14, 197375 St. Petersburg, Russia
| | - Elena Markvicheva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| |
Collapse
|
14
|
Panikkanvalappil SR, Bhagavatula SK, Deans K, Jonas O, Rashidian M, Mishra S. Enhanced Tumor Accumulation of Multimodal Magneto-Plasmonic Nanoparticles via an Implanted Micromagnet-Assisted Delivery Strategy. Adv Healthc Mater 2023; 12:e2201585. [PMID: 36213946 PMCID: PMC9840675 DOI: 10.1002/adhm.202201585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/29/2022] [Indexed: 01/18/2023]
Abstract
One of the major shortcomings of nano carriers-assisted cancer therapeutic strategies continues to be the inadequate tumor penetration and retention of systemically administered nanoformulations and its off-target toxicity. Stromal parameters-related heterogeneity in enhanced permeability and retention effect and physicochemical properties of the nanoformulations immensely contributes to their poor tumor extravasation. Herein, a novel tumor targeting strategy, where an intratumorally implanted micromagnet can significantly enhance accumulation of magneto-plasmonic nanoparticles (NPs) at the micromagnet-implanted tumor in bilateral colorectal tumor models while limiting their off-target accumulation, is demonstrated. To this end, novel multimodal gold/iron oxide NPs comprised of an array of multifunctional moieties with high therapeutic, sensing, and imaging potential are developed. It is also discovered that cancer cell targeted NPs in combination with static magnetic field can selectively induce cancer cell death. A multimodal caspase-3 nanosensor is also developed for real-time visualization of selective induction of apoptosis in cancer cells. In addition, the photothermal killing capability of these NPs in vitro is evaluated, and their potential for enhanced photothermal ablation in tissue samples is demonstrated. Building on current uses of implantable devices for therapeutic purposes, this study envisions the proposed micromagnet-assisted NPs delivery approach may be used to accelerate the clinical translation of various nanoformulations.
Collapse
Affiliation(s)
| | - Sharath K. Bhagavatula
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle Deans
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Oliver Jonas
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mohammad Rashidian
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Shruti Mishra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
15
|
Malinovskaya J, Salami R, Valikhov M, Vadekhina V, Semyonkin A, Semkina A, Abakumov M, Harel Y, Levy E, Levin T, Persky R, Chekhonin V, Lellouche JP, Melnikov P, Gelperina S. Supermagnetic Human Serum Albumin (HSA) Nanoparticles and PLGA-Based Doxorubicin Nanoformulation: A Duet for Selective Nanotherapy. Int J Mol Sci 2022; 24:ijms24010627. [PMID: 36614071 PMCID: PMC9820361 DOI: 10.3390/ijms24010627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Predicting the ability of nanoparticles (NP) to access the tumor is key to the success of chemotherapy using nanotherapeutics. In the present study, the ability of the dual NP-based theranostic system to accumulate in the tumor was evaluated in vivo using intravital microscopy (IVM) and MRI. The system consisted of model therapeutic doxorubicin-loaded poly(lactide-co-glycolide) NP (Dox-PLGA NP) and novel hybrid Ce3/4+-doped maghemite NP encapsulated within the HSA matrix (hMNP) as a supermagnetic MRI contrasting agent. Both NP types had similar sizes of ~100 nm and negative surface potentials. The level of the hMNP and PLGA NP co-distribution in the same regions of interest (ROI, ~2500 µm2) was assessed by IVM in mice bearing the 4T1-mScarlet murine mammary carcinoma at different intervals between the NP injections. In all cases, both NP types penetrated into the same tumoral/peritumoral regions by neutrophil-assisted extravasation through vascular micro- and macroleakages. The maximum tumor contrasting in MRI scans was obtained 5 h after hMNP injection/1 h after PLGA NP injection; the co-distribution level at this time reached 78%. Together with high contrasting properties of the hMNP, these data indicate that the hMNP and PLGA NPs are suitable theranostic companions. Thus, analysis of the co-distribution level appears to be a useful tool for evaluation of the dual nanoparticle theranostics, whereas assessment of the leakage areas helps to reveal the tumors potentially responsive to nanotherapeutics.
Collapse
Affiliation(s)
- Julia Malinovskaya
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
| | - Rawan Salami
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Marat Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Veronika Vadekhina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Aleksey Semyonkin
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
| | - Alevtina Semkina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Maxim Abakumov
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, Ostrovityanova ul 1, 117997 Moscow, Russia
| | - Yifat Harel
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Esthy Levy
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Tzuriel Levin
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Rachel Persky
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Vladimir Chekhonin
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Jean-Paul Lellouche
- Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
- Institute of Nanotechnology and Advanced Materials, Department of Chemistry, Faculty of Exact Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Pavel Melnikov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034 Moscow, Russia
| | - Svetlana Gelperina
- Drug Delivery Systems Laboratory, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047 Moscow, Russia
- Correspondence:
| |
Collapse
|
16
|
Schilling K, Zhai Y, Zhou Z, Zhou B, Brown E, Zhang X. High-resolution imaging of the osteogenic and angiogenic interface at the site of murine cranial bone defect repair via multiphoton microscopy. eLife 2022; 11:e83146. [PMID: 36326085 PMCID: PMC9678361 DOI: 10.7554/elife.83146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022] Open
Abstract
The spatiotemporal blood vessel formation and specification at the osteogenic and angiogenic interface of murine cranial bone defect repair were examined utilizing a high-resolution multiphoton-based imaging platform in conjunction with advanced optical techniques that allow interrogation of the oxygen microenvironment and cellular energy metabolism in living animals. Our study demonstrates the dynamic changes of vessel types, that is, arterial, venous, and capillary vessel networks at the superior and dura periosteum of cranial bone defect, suggesting a differential coupling of the vessel type with osteoblast expansion and bone tissue deposition/remodeling during repair. Employing transgenic reporter mouse models that label distinct types of vessels at the site of repair, we further show that oxygen distributions in capillary vessels at the healing site are heterogeneous as well as time- and location-dependent. The endothelial cells coupling to osteoblasts prefer glycolysis and are less sensitive to microenvironmental oxygen changes than osteoblasts. In comparison, osteoblasts utilize relatively more OxPhos and potentially consume more oxygen at the site of repair. Taken together, our study highlights the dynamics and functional significance of blood vessel types at the site of defect repair, opening up opportunities for further delineating the oxygen and metabolic microenvironment at the interface of bone tissue regeneration.
Collapse
Affiliation(s)
- Kevin Schilling
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Yuankun Zhai
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Zhuang Zhou
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| | - Bin Zhou
- Shanghai Institutes for Biological SciencesShanghaiChina
| | - Edward Brown
- Department of Biomedical Engineering, University of RochesterRochesterUnited States
| | - Xinping Zhang
- Center for Musculoskeletal Research, University of Rochester, School of Medicine and DentistryRochesterUnited States
| |
Collapse
|
17
|
Wang H, Zhou X, Wang J, Zhang X, Zhu M, Wang H. Fabrication of channeled scaffolds through polyelectrolyte complex (PEC) printed sacrificial templates for tissue formation. Bioact Mater 2022; 17:261-275. [PMID: 35386455 PMCID: PMC8965085 DOI: 10.1016/j.bioactmat.2022.01.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 12/19/2022] Open
Abstract
One of the pivotal factors that limit the clinical translation of tissue engineering is the inability to create large volume and complex three-dimensional (3D) tissues, mainly due to the lack of long-range mass transport with many current scaffolds. Here we present a simple yet robust sacrificial strategy to create hierarchical and perfusable microchannel networks within versatile scaffolds via the combination of embedded 3D printing (EB3DP), tunable polyelectrolyte complexes (PEC), and casting methods. The sacrificial templates of PEC filaments (diameter from 120 to 500 μm) with arbitrary 3D configurations were fabricated by EB3DP and then incorporated into various castable matrices (e.g., hydrogels, organic solutions, meltable polymers, etc.). Rapid dissolution of PEC templates within a 2.00 M potassium bromide aqueous solution led to the high fidelity formation of interconnected channels for free mass exchange. The efficacy of such channeled scaffolds for in vitro tissue formation was demonstrated with mouse fibroblasts, showing continuous cell proliferation and ECM deposition. Subcutaneous implantation of channeled silk fibroin (SF) scaffolds with a porosity of 76% could lead to tissue ingrowth as high as 53% in contrast to 5% for those non-channeled controls after 4 weeks. Both histological and immunofluorescence analyses demonstrated that such channeled scaffolds promoted cellularization, vascularization, and host integration along with immunoregulation.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Xiaqing Zhou
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Juan Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| | - Xinping Zhang
- School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, NY, 14642, United States
| | - Meifeng Zhu
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
- College of Life Science, Key Laboratory of Bioactive Materials, State Key Laboratory of Medicinal Chemical Biology, Xu Rongxiang Regeneration Life Science Center, Nankai University, 300071, Tianjin, PR China
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ, 07030, United States
| |
Collapse
|
18
|
Yuan B. Interstitial fluid streaming in deep tissue induced by ultrasound momentum transfer for accelerating nanoagent transport and controlling its distribution. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac88b5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/10/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Objective. This study aims to theoretically investigate the dynamics of ultrasound-induced interstitial fluid streaming and tissue recovery after ultrasound exposure for potentially accelerating nanoagent transport and controlling its distribution in tissue. Approach. Starting from fundamental equations, the dynamics of ultrasound-induced interstitial fluid streaming and tissue relaxation after an ultrasound exposure were modeled, derived and simulated. Also, both ultrasound-induced mechanical and thermal effects were considered in the models. Main results. The proposed new mechanism was named squeezing interstitial fluid via transfer of ultrasound momentum (SIF-TUM). It means that an ultrasound beam can squeeze the tissue in a small focal volume from all the directions, and generate a macroscopic streaming of interstitial fluid and a compression of tissue solid matrix. After the ultrasound is turned off, the solid matrix will recover and can generate a backflow. Rather than the ultrasound pressure itself or intensity, the streaming velocity is determined by the dot product of the ultrasound pressure gradient and its conjugate. Tissue and nanoagent properties also affect the streaming and recovery velocities. Significance. The mobility of therapeutic or diagnostic agents, such as drugs, drug carriers, or imaging contrast agents, in the interstitial space of many diseased tissues, such as tumors, is usually extremely low because of the inefficiency of the natural transport mechanisms. Therefore, the interstitial space is one of the major barriers hindering agent deliveries. The ability to externally accelerate agent transport and control its distribution is highly desirable. Potentially, SIF-TUM can be a powerful technology to accelerate agent transport in deep tissue and control the distribution if appropriate parameters are selected.
Collapse
|
19
|
O'Melia MJ, Rohner NA, Thomas SN. Tumor Vascular Remodeling Affects Molecular Dissemination to Lymph Node and Systemic Leukocytes. Tissue Eng Part A 2022; 28:781-794. [PMID: 35442085 PMCID: PMC9508451 DOI: 10.1089/ten.tea.2022.0020] [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: 01/31/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Angiogenic and lymphangiogenic remodeling has long been accepted as a hallmark of cancer development and progression; however, the impacts of this remodeling on immunological responses, which are paramount to the responses to immunotherapeutic treatments, are underexplored. As immunotherapies represent one of the most promising new classes of cancer therapy, in this study, we explore the effects of angiogenic and lymphangiogenic normalization on dissemination of molecules injected into the tumor microenvironment to immune cells in lymph nodes draining the tumor as well as in systemically distributed tissues. A system of fluorescent tracers, size-matched to biomolecules of interest, was implemented to track different mechanisms of tumor transport and access to immune cells. This revealed that the presence of a tumor, and either angiogenic or lymphangiogenic remodeling, altered local retention of model biomolecules, trended toward normalizing dissemination to systemic organs, and modified access to lymph node-resident immune cells in manners dependent on mechanism of transport. More specifically, active cell migration by skin-derived antigen presenting cells was enhanced by both the presence of a tumor and lymphangiogenic normalization, while both angiogenic and lymphangiogenic normalization restored patterns of immune cell access to passively draining species. As a whole, this work uncovers the potential ramifications of tumor-induced angiogenesis and lymphangiogenesis, along with impacts of interrogation into these pathways, on access of tumor-derived species to immune cells. Impact Statement Angiogenic and lymphangiogenic normalization strategies have been utilized clinically to interrogate tumor vasculature with some success. In the age of immunotherapy, the impacts of these therapeutic interventions on immune remodeling are unclear. This work utilizes mouse models of angiogenic and lymphangiogenic normalization, along with a system of fluorescently tagged tracers, to uncover the impacts of angiogenesis and lymphangiogenesis on access of tumor-derived species to immune cell subsets within various organs.
Collapse
Affiliation(s)
- Meghan J. O'Melia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
| | - Nathan A. Rohner
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Susan Napier Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
20
|
Wu C, Zhang Y, Wei X, Li N, Huang H, Xie Z, Zhang H, Yang G, Li M, Li T, Yang H, Li S, Qin X, Liu Y. Tumor Homing-Penetrating and Nanoenzyme-Augmented 2D Phototheranostics Against Hypoxic Solid Tumors. Acta Biomater 2022; 150:391-401. [DOI: 10.1016/j.actbio.2022.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/15/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
|
21
|
Desai P, Rimal R, Florea A, Gumerov RA, Santi M, Sorokina AS, Sahnoun SEM, Fischer T, Mottaghy FM, Morgenroth A, Mourran A, Potemkin II, Möller M, Singh S. Tuning the Elasticity of Nanogels Improves Their Circulation Time by Evading Immune Cells. Angew Chem Int Ed Engl 2022; 61:e202116653. [PMID: 35274425 PMCID: PMC9325431 DOI: 10.1002/anie.202116653] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Indexed: 12/22/2022]
Abstract
Peptide receptor radionuclide therapy is used to treat solid tumors by locally delivering radiation. However, due to nephro‐ and hepato‐toxicity, it is limited by its dosage. To amplify radiation damage to tumor cells, radiolabeled nanogels can be used. We show that by tuning the mechanical properties of nanogels significant enhancement in circulation half‐life of the gel could be achieved. We demonstrate why and how small changes in the mechanical properties of the nanogels influence its cellular fate. Nanogels with a storage modulus of 37 kPa were minimally phagocytosed by monocytes and macrophages compared to nanogels with 93 kPa modulus. Using PET/CT a significant difference in the blood circulation time of the nanogels was shown. Computer simulations affirmed the results and predicted the mechanism of cellular uptake of the nanogels. Altogether, this work emphasizes the important role of elasticity even for particles that are inherently soft such as nano‐ or microgels.
Collapse
Affiliation(s)
- Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Rahul Rimal
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Alexandru Florea
- Department of Nuclear Medicine University Hospital RWTH Aachen Pauwelstraße 30 52074 Aachen Germany
- Department of Radiology and Nuclear Medicine School for Cardiovascular Diseases (CARIM) and School for Oncology (GROW) Maastricht University 6229 HX Maastricht The Netherlands
| | - Rustam A. Gumerov
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
- Physics Department Lomonosov Moscow State University Leninskie Gory 1–2 119991 Moscow Russian Federation
| | - Marta Santi
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Anastasia S. Sorokina
- Physics Department Lomonosov Moscow State University Leninskie Gory 1–2 119991 Moscow Russian Federation
| | - Sabri E. M. Sahnoun
- Department of Nuclear Medicine University Hospital RWTH Aachen Pauwelstraße 30 52074 Aachen Germany
| | - Thorsten Fischer
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Felix M. Mottaghy
- Department of Nuclear Medicine University Hospital RWTH Aachen Pauwelstraße 30 52074 Aachen Germany
- Department of Radiology and Nuclear Medicine School for Cardiovascular Diseases (CARIM) and School for Oncology (GROW) Maastricht University 6229 HX Maastricht The Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine University Hospital RWTH Aachen Pauwelstraße 30 52074 Aachen Germany
| | - Ahmed Mourran
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Igor I. Potemkin
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
- Physics Department Lomonosov Moscow State University Leninskie Gory 1–2 119991 Moscow Russian Federation
- National Research South Ural State University Chelyabinsk 454080 Russian Federation
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
| | - Smriti Singh
- DWI Leibniz Institute for Interactive Materials e.V RWTH Aachen University Forckenbeckstrasse 50 52074 Aachen Germany
- Max Planck Institute for Medical Research (MPImF) Jahnstrasse 29 69120 Heidelberg Germany
| |
Collapse
|
22
|
Desai P, Rimal R, Florea A, Gumerov RA, Santi M, Sorokina AS, Sahnoun SEM, Fischer T, Mottaghy FM, Morgenroth A, Mourran A, Potemkin II, Möller M, Singh S. Tuning the Elasticity of Nanogels Improves their Circulation Time by Evading Immune Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202116653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Prachi Desai
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular Chemistry Aachen GERMANY
| | - Rahul Rimal
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular chemistry Aachen GERMANY
| | - Alexandru Florea
- Uniklinik RWTH Aachen: Universitatsklinikum Aachen Nuclear Medicine GERMANY
| | - Rustam A. Gumerov
- Lomonosov Moscow State University: Moskovskij gosudarstvennyj universitet imeni M V Lomonosova Physics RUSSIAN FEDERATION
| | - Marta Santi
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular Chemistry GERMANY
| | - Anastasia S. Sorokina
- Lomonosov Moscow State University: Moskovskij gosudarstvennyj universitet imeni M V Lomonosova Physics RUSSIAN FEDERATION
| | | | - Thorsten Fischer
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular Chemistry GERMANY
| | - Felix M. Mottaghy
- Uniklinik RWTH Aachen: Universitatsklinikum Aachen Nuclear Medicine GERMANY
| | | | - Ahmed Mourran
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular chemistry GERMANY
| | - Igor I. Potemkin
- Lomonosov Moscow State University: Moskovskij gosudarstvennyj universitet imeni M V Lomonosova Physics RUSSIAN FEDERATION
| | - Martin Möller
- DWI-Leibniz-Institut für Interaktive Materialien: DWI-Leibniz-Institut fur Interaktive Materialien Macromolecular Chemistry GERMANY
| | - Smriti Singh
- Max-Planck-Institute for Medical Research: Max-Planck-Institut fur medizinische Forschung Cellular Biophysics Jahnstr. 29 Heidelberg GERMANY
| |
Collapse
|
23
|
Della Sala F, Silvestri T, Borzacchiello A, Mayol L, Ambrosio L, Biondi M. Hyaluronan-coated nanoparticles for active tumor targeting: Influence of polysaccharide molecular weight on cell uptake. Colloids Surf B Biointerfaces 2021; 210:112240. [PMID: 34864635 DOI: 10.1016/j.colsurfb.2021.112240] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/29/2021] [Accepted: 11/18/2021] [Indexed: 11/20/2022]
Abstract
Here we aimed to correlate different molecular weights of hyaluronic acid (HA), 200, 800 and 1437 kDa, used to decorate poly(lactic-co-glycolic acid) (PLGA)-based nanoparticles (NPs), to their cell uptakes. NP internalization kinetics in CD44-overexpressing breast carcinoma cells were quantified, using healthy fibroblast cells as reference. Actually, NP uptake and selectivity by tumor cells were maximized for NPs HA 800 kDa, while being minimum for NPs HA1400 kDa. This unexpected result could be explained considering that the interaction between NPs and tumor cells is dictated by rearrangement and conformation of that segment of HA chain that actually protrudes from the NPs. Overall, results obtained in this work point at how HA molecular weight, is pivotal project parameter in NP formulation to promote active targeting in the CD44 overexpressing cancer cells.
Collapse
Affiliation(s)
- Francesca Della Sala
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy
| | - Teresa Silvestri
- Dipartimento di Farmacia, Università di Napoli Federico II, Via Domenico Montesano 49, Napoli, Italy
| | - Assunta Borzacchiello
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy.
| | - Laura Mayol
- Dipartimento di Scienze Biomediche Avanzate, scuola di Medicina e Chirurgia, Università degli studi di Napoli Federico II, Via Domenico Montesano 49, Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali (CRIB), Università di Napoli Federico II, Piazzale Tecchio 80, Napoli, Italy.
| | - Luigi Ambrosio
- Istituto per i Polimeri, Compositi e Biomateriali, Consiglio Nazionale delle Ricerche (IPCB-CNR), Viale J.F. Kennedy 54, Napoli, Italy
| | - Marco Biondi
- Dipartimento di Farmacia, Università di Napoli Federico II, Via Domenico Montesano 49, Napoli, Italy; Centro di Ricerca Interdipartimentale sui Biomateriali (CRIB), Università di Napoli Federico II, Piazzale Tecchio 80, Napoli, Italy
| |
Collapse
|
24
|
Cai F, Li S, Huang H, Iqbal J, Wang C, Jiang X. Green synthesis of gold nanoparticles for immune response regulation: Mechanisms, applications, and perspectives. J Biomed Mater Res A 2021; 110:424-442. [PMID: 34331516 DOI: 10.1002/jbm.a.37281] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
Immune responses are involved in the pathogenesis of many diseases, including cancer, autoimmune diseases, and chronic inflammation. These responses are attributed to immune cells that produce cytokines, mediate cytotoxicity, and synthesize antibodies. Gold nanoparticles (GNPs) are novel agents that intervene with immune responses because of their unique physical-chemical properties. In particular, GNPs enhance anti-tumour activity during immunotherapy and eliminate excessive inflammation in autoimmune diseases. However, GNPs synthesized by conventional methods are toxic to living organisms. Green biosynthesis provides a safe and eco-friendly method to obtain GNPs from microbes or plant extracts. In this review, we describe several patterns for green GNP biosynthesis. The applications of GNPs to target immune cells and modulate the immune response are summarized. In particular, we elaborate on how GNPs regulate innate immunity and adaptive immunity, including inflammatory signaling and immune cell differentiation. Finally, perspectives and challenges in utilizing green biosynthesized GNPs for novel therapeutic approaches are discussed.
Collapse
Affiliation(s)
- Feiyang Cai
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China.,School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Huang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Javed Iqbal
- Department of Botany, Bacha Khan University, Charsadda, Khyber Pakhtunkhwa, Pakistan
| | - Canran Wang
- School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Jiang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
25
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
26
|
Patel JP, Spiller SE, Barker ED. Drug penetration in pediatric brain tumors: Challenges and opportunities. Pediatr Blood Cancer 2021; 68:e28983. [PMID: 33719183 DOI: 10.1002/pbc.28983] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/23/2022]
Abstract
Larger clinical trial enrollments and a greater understanding of biological heterogeneity have led to improved survival rates for children diagnosed with brain tumors in the last 50 years. However, reducing long-term morbidities and improving survival rates of high-risk tumors remain major challenges. Chemotherapy can reduce tumor burden, but effective drug penetration at the tumor site is limited by barriers in the route of drug administration and within the tumor microenvironment. Bioavailability of drugs is impeded by the blood-brain barrier, plasma protein binding, and structural components by the tumor including the matrix and vasculature contributing to increased interstitial fluid pressure, hypoxia, and acidity. Designing drug delivery systems to circumvent these barriers could lead to improved drug penetration at the tumor site and reduce adverse systemic side effects. In this review, we expand on how systemic and local barriers limit drug penetration and present potential methods to enhance drug penetration in pediatric brain tumors.
Collapse
Affiliation(s)
- Jenny P Patel
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| | - Susan E Spiller
- Pediatric Hematology/Oncology, East Tennessee Children's Hospital, Knoxville, Tennessee
| | - Elizabeth D Barker
- Department of Mechanical, Aerospace, and Biomedical Engineering, The University of Tennessee at Knoxville, Knoxville, Tennessee
| |
Collapse
|
27
|
Oh HJ, Kim J, Kim H, Choi N, Chung S. Microfluidic Reconstitution of Tumor Microenvironment for Nanomedical Applications. Adv Healthc Mater 2021; 10:e2002122. [PMID: 33576178 DOI: 10.1002/adhm.202002122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Indexed: 12/17/2022]
Abstract
Nanoparticles have an extensive range of diagnostic and therapeutic applications in cancer treatment. However, their current clinical translation is slow, mainly due to the failure to develop preclinical evaluation techniques that can draw similar conclusions to clinical outcomes by adequately mimicking nanoparticle behavior in complicated tumor microenvironments (TMEs). Microfluidic methods offer significant advantages over conventional in vitro methods to resolve these challenges by recapitulating physiological cues of the TME such as the extracellular matrix, shear stress, interstitial flow, soluble factors, oxygen, and nutrient gradients. The methods are capable of de-coupling microenvironmental features, spatiotemporal controlling of experimental sequences, and high throughput readouts in situ. This progress report highlights the recent achievements of microfluidic models to reconstitute the physiological microenvironment, especially for nanomedical tools for cancer treatment.
Collapse
Affiliation(s)
- Hyun Jeong Oh
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Jaehoon Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Hyunho Kim
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems Brain Science Institute Korea Institute of Science and Technology (KIST) Seoul 02792 Republic of Korea
- Division of Bio‐Medical Science & Technology KIST School Korea University of Science and Technology (UST) Seoul 34113 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering Korea University Seoul 02841 Republic of Korea
- KU‐KIST Graduate School of Converging Science and Technology Korea University Seoul 02841 Republic of Korea
| |
Collapse
|
28
|
Yu Z, Jiang F, Hu C, Tang B. Functionalized nanoprobes for in situ detection of telomerase. Chem Commun (Camb) 2021; 57:3736-3748. [PMID: 33876119 DOI: 10.1039/d0cc08412c] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Telomerase, a special ribonucleoprotein reverse transcriptase, can maintain the length and stability of telomeres and plays an important role in cell proliferation and differentiation. Due to the distinguishable expression level in normal cells and cancer cells, telomerase has become an important biomarker for cancer diagnosis and prognosis evaluation. Despite major breakthroughs in the field of telomerase detection, the extracts in the cell lysate are still the first choice as the analyte nevertheless, which will bring serious inaccuracies compared with the real intracellular activity. With the development of nanotechnology and nanomaterials, extraordinary progress has been made in telomerase detection by employing different versatile nanoprobes. In this review, we list the superiority of nanoprobes and systematically summarize the applications of nanoprobes in telomerase detection from the aspects of various nanomaterials and discuss the current challenges and potential trends in the future design of nanoprobes.
Collapse
Affiliation(s)
- Zhengze Yu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China.
| | | | | | | |
Collapse
|
29
|
Zheng Y, Fang X, Yang Y, Wang C. Peptide-directed delivery of drug-loaded nanocarriers targeting CD36 overexpressing cells. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2020.125970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release 2021; 329:624-644. [PMID: 33010333 PMCID: PMC8082750 DOI: 10.1016/j.jconrel.2020.09.055] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Clinically efficacious medication in anticancer therapy has been successfully designed with liposome-based nanomedicine. The liposomal formulation in cancer drug delivery can be facilitated with a functionalized peptide that mediates the specific drug delivery opportunities with increased drug penetrability, specific accumulation in the targeted site, and enhanced therapeutic efficacy. This review aims to focus on recent advances in peptide-functionalized liposomal formulation techniques in cancer diagnosis and treatment regarding recently published literature. It also will highlight different aspects of novel liposomal formulation techniques that incorporate surface functionalization with peptides for better anticancer effect and current challenges in peptide-functionalized liposomal drug formulation.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
31
|
Wang W, Zhou C. A Journey of Nanomotors for Targeted Cancer Therapy: Principles, Challenges, and a Critical Review of the State-of-the-Art. Adv Healthc Mater 2021; 10:e2001236. [PMID: 33111501 DOI: 10.1002/adhm.202001236] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Indexed: 12/11/2022]
Abstract
A nanomotor is a miniaturized device that converts energy stored in the environment into mechanical motion. The last two decades have witnessed a surge of research interests in the biomedical applications of nanomotors, but little clinical translation. To accelerate this process, targeted cancer therapy is used as an example to describe a "survive, locate, operate, and terminate" (SLOT) mission of a nanomotor, where it must 1) survive in the unfriendly in vivo environment, 2) locate its target as well as be located by human operators, 3) carry out specific operations, and 4) terminate after the mission is completed. Along this journey, the challenges presented to a nanomotor, including to power, navigate, steer, target, release, control, image, and communicate are discussed, and how state-of-the-art nanomotors meet or fall short of these requirements is critically reviewed. These discussions are then condensed into a table for easy reference. In particular, it is argued that chemically powered nanomotors are intrinsically ill-positioned for targeted cancer therapy, while nanomotors powered by magnetic fields or ultrasound show more promises. Following this argument, a tentative nanomotor design is then presented in the end to conform to the SLOT guideline, and to inspire practical, functional nanorobots that are yet to come.
Collapse
Affiliation(s)
- Wei Wang
- School of Materials Science and Engineering Harbin Institute of Technology (Shenzhen) Shenzhen 518055 China
| | - Chao Zhou
- School of Materials Science and Engineering Harbin Institute of Technology (Shenzhen) Shenzhen 518055 China
| |
Collapse
|
32
|
Ma T, Xia T. Nanoparticle-Based Activatable Probes for Bioimaging. Adv Biol (Weinh) 2021; 5:e2000193. [PMID: 33724732 PMCID: PMC7966733 DOI: 10.1002/adbi.202000193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022]
Abstract
Molecular imaging can provide functional and molecular information at the cellular or subcellular level in vivo in a noninvasive manner. Activatable nanoprobes that can react to the surrounding physiological environment or biomarkers are appealing agents to improve the efficacy, specificity, and sensitivity of molecular imaging. The physiological parameters, including redox status, pH, presence of enzymes, and hypoxia, can be designed as the stimuli of the activatable probes. However, the success rate of imaging nanoprobes for clinical translation is low. Herein, the recent advances in nanoparticle-based activatable imaging probes are critically reviewed. In addition, the challenges for clinical translation of these nanoprobes are also discussed in this review.
Collapse
Affiliation(s)
- Tiancong Ma
- Division of Nanomedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1772, USA
- Department of Environmental Health Sciences, Jonathan and Karin Fielding School of Public Health, University of California, Los Angeles, California 90095-1772, USA
| | - Tian Xia
- Division of Nanomedicine, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095-1772, USA
| |
Collapse
|
33
|
Ali HR, Selim SA, Aili D. Effects of macrophage polarization on gold nanoparticle-assisted plasmonic photothermal therapy. RSC Adv 2021; 11:25047-25056. [PMID: 35481041 PMCID: PMC9037012 DOI: 10.1039/d1ra03671h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/03/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor associated macrophages (TAM) are key pathogenic factors in neoplastic diseases. They are known to have plasticity and can polarize into two opposing phenotypes, including the tumoricidal M1 and the protumoral M2 phenotypes with high prevalence of M2-phentoypes in patients with poor prognosis. Strategies for targeting M2-TAM may consequently increase the efficacy of therapeutic strategies for cancer treatment. Gold nanorod-assisted plasmonic photothermal therapy (PPTT) has emerged as a promising treatment for cancer but the effects of macrophage polarization parameters in the performance of this new treatment modality is still unknown. Herein, human monocytic THP-1 cells were polarized into two opposite phenotypic macrophages (M1-TAM and M2-TAM) and their response to PPTT was examined. M2-TAM exhibits a three-fold increase in AuNP uptake compared to M1-TAM. Laser irradiation results in selective killing of pro-tumoral M2-TAM after treatment with AuNPs with limited effects on anti-tumoral M1-TAM. A positive correlation between the expression of CD206 marker and the AuNP uptake may indicate the role of CD206 in facilitating AuNP uptake. Our findings also suggest that the differences in AuNP avidity and uptake between the M1-TAM and M2-TAM phenotypes may be the rationale behind the effectiveness of PPTT in the treatment of solid tumors. A preferential uptake of gold nanoparticles by macrophages with a protumoral M2 phenotype result in efficient killing upon laser irradiation while keeping M1 phenotypes relatively undamaged.![]()
Collapse
Affiliation(s)
- Hala R. Ali
- Department of Bacteriology and Immunology
- Animal Health Research Institute (AHRI)
- Agriculture Research Center (ARC)
- Egypt
| | - Salah A. Selim
- Department of Microbiology
- Faculty of Veterinary Medicine
- Cairo University
- Giza
- Egypt
| | - Daniel Aili
- Laboratory of Molecular Materials
- Division of Biophysics and Bioengineering
- Department of Physics, Chemistry and Biology
- Linköping University
- SE-581 83 Linköping
| |
Collapse
|
34
|
Sandha KK, Shukla MK, Gupta PN. Recent Advances in Strategies for Extracellular Matrix Degradation and Synthesis Inhibition for Improved Therapy of Solid Tumors. Curr Pharm Des 2020; 26:5456-5467. [DOI: 10.2174/1381612826666200728141601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/27/2020] [Indexed: 01/04/2023]
Abstract
Despite a great deal of efforts made by researchers and the advances in the technology, the treatment of
cancer is very challenging. Significant advances in the field of cancer therapeutics have been made but due to the
complexity of solid tumor microenvironment, specially their dense extracellular matrix (which makes the conditions
favorable for cancer growth, metastasis and acts as a barrier to the chemotherapeutic drugs as well as
nanomedicine), the treatment of solid tumors is difficult. Overexpression of extracellular matrix components such
as collagen, hyaluronan and proteoglycans in solid tumor leads to high interstitial fluid pressure, hypoxia, vascular
collapse and poor perfusion which hinder the diffusion and convection of the drugs into the tumor tissue. This
leads to the emergence of drug resistance and poor antitumor efficacy of chemotherapeutics. A number of approaches
are being investigated in order to modulate this barrier for improved outcome of cancer chemotherapy.
In this review, recent advances in the various approaches for the modulation of the extracellular matrix barrier of
the solid tumor are covered and significant findings are discussed in an attempt to facilitate more investigations in
this potential area to normalize the tumor extracellular matrix for improving drug exposure to solid tumor.
Collapse
Affiliation(s)
- Kamalpreet Kaur Sandha
- PK-PD Toxicology & Formulation Division, CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu, J&K, 180001, India
| | - Monu Kumar Shukla
- PK-PD Toxicology & Formulation Division, CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu, J&K, 180001, India
| | - Prem N. Gupta
- PK-PD Toxicology & Formulation Division, CSIR- Indian Institute of Integrative Medicine, Canal Road, Jammu, J&K, 180001, India
| |
Collapse
|
35
|
Wu S, Lin X. Trials in developing a nanoscale material for extravascular contrast-enhanced ultrasound targeting hepatocellular carcinoma. PeerJ 2020; 8:e10403. [PMID: 33354418 PMCID: PMC7727372 DOI: 10.7717/peerj.10403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/31/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Medical imaging is an important approach for the diagnosis of hepatocellular carcinoma (HCC), a common life threaten disease, however, the diagnostic efficiency is still not optimal. Developing a novel method to improve diagnosis is necessary. The aim of this project was to formulate a material that can combine with GPC3 of HCC for targeted enhanced ultrasound. METHODS A material of sulfur hexafluoride (SF6) filled liposome microbubbles and conjugated with synthesized peptide (LSPMbs) was prepared and assessed in vitro and vivo. Liposome microbubbles were made of DPPC, DPPG, DSPE-PEG2000,and SF6, using thin film method to form shell, followed filling SF6, and conjugating peptide. A carbodiimide method was used for covalent conjugation of peptide to LSMbs. RESULTS The prepared LSPMbs appeared round shaped, with size of 380.9 ± 176.5 nm, and Zeta potential of -51.4 ± 10.4mV. LSPMbs showed high affinity to Huh-7 cells in vitro, presented good enhanced ultrasound effects, did not show cytotoxicity, and did not exhibit targeted fluorescence and enhanced ultrasound in animal xenograft tumors. CONCLUSION Extravascular contrast-enhanced ultrasound targeted GPC3 on HCC may not be realized, and the reason may be that targeted contrast agents of microbubbles are hard to access and accumulate in the tumor stroma and matrix.
Collapse
Affiliation(s)
- Size Wu
- Department of Ultrasound, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Xiyuan Lin
- Department of Emergency Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| |
Collapse
|
36
|
Wang P, Wu M, Li A, Ye X, Li C, Xu D. Diagnostic Value of Contrast-Enhanced Ultrasound for Differential Diagnosis of Malignant and Benign Soft Tissue Masses: A Meta-Analysis. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:3179-3187. [PMID: 32907771 DOI: 10.1016/j.ultrasmedbio.2020.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/22/2020] [Accepted: 08/07/2020] [Indexed: 06/11/2023]
Abstract
This meta-analysis was aimed at investigating the value of using contrast-enhanced ultrasound (CEUS) in the differential diagnosis of benign and malignant soft tissue masses (STMs). Relevant studies published before March 24, 2020 were identified through a comprehensive search of PubMed, Ovid, Cochrane and Web of Science. According to the inclusion criteria, five studies were selected comprising 746 patients. In the differential diagnosis of benign and malignant STMs, the pooled sensitivity and specificity of CEUS were 76% (95% confidence interval [CI]: 71%-81%; heterogeneity [I2] = 74.5%) and 67% (95% CI: 62%-71%; I2 = 36.5%), respectively. The diagnostic odds ratio was 7.37 (95% CI: 3.78%-14.35; I2 = 66.6%). The overall area under the curve was 0.77 (standard error: 0.0392). Subgroup analysis revealed that different index tests of CEUS resulted in different diagnostic performance. Importantly, CEUS is an effective method for the differential diagnosis between benign and malignant STMs.
Collapse
Affiliation(s)
- Pingping Wang
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mengjie Wu
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ao Li
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhua Ye
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cuiying Li
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Di Xu
- Department of Ultrasound, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
37
|
Zhang Y, Dong Y, Fu H, Huang H, Wu Z, Zhao M, Yang X, Guo Q, Duan Y, Sun Y. Multifunctional tumor-targeted PLGA nanoparticles delivering Pt(IV)/siBIRC5 for US/MRI imaging and overcoming ovarian cancer resistance. Biomaterials 2020; 269:120478. [PMID: 33213862 DOI: 10.1016/j.biomaterials.2020.120478] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Cisplatin (Pt(II)) resistance is an important factor in the high mortality rates of ovarian cancer. Herein, we synthesized multifunctional tumor-targeted poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs-cRGD) for monitoring therapeutic effects by dual-mode imaging and overcoming cisplatin resistance. Uniformly sized NPs-cRGD demonstrated controlled and sustained release of drugs and genes, excellent gene loading and gene protection capacity, good storage stability and no serum-induced aggregation in vitro. NPs-cRGD demonstrated clear, targeting and prolonged ultrasound imaging and magnetic resonance imaging (MRI) in vivo. The targeting of NPs-cRGD combined with ultrasound facilitated nanoparticle penetrattion into cells; entry was time-dependent. NPs-cRGD escaped from lysosomes, thereby preventing siBIRC5 degradation, which enabled siBIRC5 to efficiently inhibit the antiapoptosis effects of BIRC5 in SKO3-DDP to overcome the antiapoptosis properties of resistant cells. Furthermore, Pt(IV) in NPs-cRGD exhausted glutathione (GSH), thereby increasing drug accumulation to effectively increase Pt(II) levels. The subsequent combination of Pt(II) with DNA prevented the expressions of genes and upregulated the expression of p53 to induce the mitochondria apoptosis pathway. The reduced GSH activity and the generation of Pt(II) further promoted high levels of reactive oxygen species (ROS) to induce cell apoptosis. Therefore, NPs-cRGD with ultrasound promoted the apoptosis of resistant ovarian cancer cells by multiple mechanisms, including increased cellular drug accumulation, reversed antiapoptotic effects by siBIRC5, and enhanced ROS levels. In a tumor-bearing nude mice model, NPs-cRGD with US demonstrated excellent tumor-targeting, high efficiency tumor inhibition and low systemic toxicity. Therefore, NPs-cRGD provides a means to monitor treatment processes and can be combined with ultrasound treatment to overcome ovarian cancer resistance in vitro and in vivo.
Collapse
Affiliation(s)
- Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Zhihua Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Meng Zhao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xupeng Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianqian Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
38
|
Alijaj N, Moutel S, Gouveia ZL, Gray M, Roveri M, Dzhumashev D, Weber F, Meier G, Luciani P, Rössler JK, Schäfer BW, Perez F, Bernasconi M. Novel FGFR4-Targeting Single-Domain Antibodies for Multiple Targeted Therapies against Rhabdomyosarcoma. Cancers (Basel) 2020; 12:cancers12113313. [PMID: 33182650 PMCID: PMC7696840 DOI: 10.3390/cancers12113313] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 01/08/2023] Open
Abstract
Simple Summary Rhabdomyosarcoma (RMS) accounts for more than 50% of all soft tissue sarcomas in childhood and adolescence. Despite progress and intensified multimodality treatment, prognoses are extremely poor with an overall survival rate of approximately 20% in the advanced stage. Therefore, there is an urgent need for targeted treatment options to improve overall survival rates, and to limit long-term side effects. The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in RMS and other tumors as well. The goal of this work was to select FGFR4 specific single-domain antibodies (sdAb) and to develop FGFR4-targeted therapies. We could show that FGFR4-targeted liposomes have the potential to deliver drugs specifically to FGFR4-positive tumor cells and that chimeric antigen receptor T cells built with the selected antibodies can kill specifically FGFR4-expressing RMS cells. Abstract The fibroblast growth factor receptor 4 (FGFR4) is overexpressed in rhabdomyosarcoma (RMS) and represents a promising target for treatments based on specific and efficient antibodies. Despite progress, there is an urgent need for targeted treatment options to improve survival rates, and to limit long-term side effects. From phage display libraries we selected FGFR4-specific single-domain antibodies (sdAb) binding to recombinant FGFR4 and validated them by flow cytometry, surface plasmon resonance, and fluorescence microscopy. The specificity of the selected sdAb was verified on FGFR4-wild type and FGFR4-knock out cells. FGFR4-sdAb were used to decorate vincristine-loaded liposomes and to generate chimeric antigen receptor (CAR) T cells. First, incubation of RMS cells with FGFR4-sdAb revealed that FGFR4-sdAb can block FGF19-FGFR4 signaling via the MAPK pathway and could therefore serve as therapeutics for FGFR4-dependent cancers. Second, FGFR4-targeted vincristine-loaded liposomes bound specifically to RMS cells and were internalized by the receptor, demonstrating the potential for active drug delivery to the tumor. Third, FGFR4-CAR T cells, generated with one sdAb candidate, demonstrated strong and specific cytotoxicity against FGFR4 expressing RMS cells. We selected novel FGFR4-sdAb with high specificity and nano- to picomolar affinities for FGFR4 which have the potential to enable multiple FGFR4-targeted cancer therapy approaches.
Collapse
Affiliation(s)
- Nagjie Alijaj
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Sandrine Moutel
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Recombinant Antibody Platform (TAb-IP), Institut Curie, 75248 Paris, France
| | - Zelia L. Gouveia
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Honing Biosciences, 75004 Paris, France
| | - Maxim Gray
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Maurizio Roveri
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Dzhangar Dzhumashev
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Florian Weber
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Gianmarco Meier
- Institute of Medical Microbiology, University of Zurich, 8006 Zurich, Switzerland;
| | - Paola Luciani
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland; (F.W.); (P.L.)
| | - Jochen K. Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Beat W. Schäfer
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
| | - Franck Perez
- Institut Curie, PSL Research University, CNRS UMR144, 75248 Paris, France; (S.M.); (Z.L.G.)
- Correspondence: (F.P.); (M.B.)
| | - Michele Bernasconi
- Department of Oncology, Children’s Research Center, University Children’s Hospital Zurich, 8032 Zurich, Switzerland; (N.A.); (M.G.); (M.R.); (B.W.S.)
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland; (D.D.); (J.K.R.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Correspondence: (F.P.); (M.B.)
| |
Collapse
|
39
|
Interstitial Hypertension Suppresses Escape of Human Breast Tumor Cells Via Convection of Interstitial Fluid. Cell Mol Bioeng 2020; 14:147-159. [PMID: 33868497 DOI: 10.1007/s12195-020-00661-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 10/26/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction Interstitial hypertension, a rise in interstitial fluid pressure, is a common feature of many solid tumors as they progress to an invasive state. It is currently unclear whether this elevated pressure alters the probability that tumor cells eventually escape into a neighboring blood or lymphatic vessel. Methods In this study, we analyze the escape of MDA-MB-231 human breast tumor cells from a ~3-mm-long preformed aggregate into a 120-μm-diameter empty cavity in a micromolded type I collagen gel. The "micro-tumors" were located within ~300 μm of one or two cavities. Pressures of ~0.65 cm H2O were applied only to the tumor ("interstitial hypertension") or to its adjacent cavity. Results This work shows that interstitial hypertension suppresses escape into the adjacent cavity, but not because tumor cells respond directly to the pressure profile. Instead, hypertension alters the chemical microenvironment at the tumor margin to one that hampers escape. Administration of tumor interstitial fluid phenocopies the effects of hypertension. Conclusions This work uncovers a link between tumor pressure, interstitial flow, and tumor cell escape in MDA-MB-231 cells, and suggests that interstitial hypertension serves to hinder further progression to metastatic escape. Electronic Supplementary Material The online version of this article (10.1007/s12195-020-00661-w) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Whitener R, Mosley RJ, Wower J, Byrne ME. Nucleic acid biohybrid nanocarriers with high-therapeutic payload and controllable extended release of daunomycin for cancer therapy. J Biomed Mater Res A 2020; 109:1256-1265. [PMID: 33047466 DOI: 10.1002/jbm.a.37119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 11/07/2022]
Abstract
We have developed a novel, nanosized drug carrier with high-therapeutic payload, controllable release, and the potential for active tumor targeting. It consists of a 15 nm gold nanoparticle with dense surface loading of DNA duplexes. We utilize the natural intercalating behavior of daunomycin to load the drug between DNA base pairs. We obtained a high-therapeutic payload of >1,000 drug molecules per gold nanoparticle (AuNP), one of the highest loadings reported in literature to date. We have engineered unique DNA sequences to control release of daunomycin for over 48 hr and show higher cell death compared to equivalent concentrations of free daunomycin. We have also explored cell internalization mechanisms to identify the pathways by which our gold nanoparticles enter the cell. This nanocarrier is in the ideal size range of 16-100 nm in diameter to utilize the enhanced permeability and retention effect for passive targeting to tumors. Our AuNP platform is effective as a therapeutic drug delivery device and can easily incorporate any aptamer of choice through complementary base pairing. Our work has produced an innovative nanoscale drug-delivery platform potentially leading to personalized cancer therapies through careful selection of aptamers and an adjustable drug release profile.
Collapse
Affiliation(s)
- Ricky Whitener
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Robert J Mosley
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| | - Jacek Wower
- RNA Biochemistry Laboratories, Department of Animal Sciences, Auburn University, Auburn, Alabama, USA
| | - Mark Edward Byrne
- Biomimetic and Biohybrid Materials, Biomedical Devices, and Drug Delivery Laboratories, Department of Biomedical Engineering, Rowan University, Glassboro, New Jersey, USA
| |
Collapse
|
41
|
Electrostatic driven transport enhances penetration of positively charged peptide surfaces through tumor extracellular matrix. Acta Biomater 2020; 113:240-251. [PMID: 32428687 DOI: 10.1016/j.actbio.2020.04.051] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022]
Abstract
Drug carriers achieve poor and heterogeneous distribution within solid tumors due to limited transport through the tumor extracellular matrix (ECM). The tumor ECM forms a net negatively charged network that interacts with and hinders the transport of molecules in part due to electrostatic interactions. Traditionally, the surfaces of drug delivery systems are passivated to minimize these interactions, but the mechanism of how charge interactions impact transport and penetration within the tumor microenvironment (TME) is not well understood. Here, we used T7 bacteriophage as a model biological nanoparticle to display peptides of different charges on its surface and elucidate how charge-based binding drives transport, uptake, and retention within tumor tissue. In contrast to current studies with neutrally charged surfaces, we discovered that a positively charged peptide displayed on T7 enhanced its penetration through a tumor-like ECM when compared to neutrally and negatively charged peptides. The positively charged peptide displayed on T7 facilitated weak and reversible binding with the TME to achieve Donnan partitioning and deep penetration into ex vivo tumor tissue. Additionally, the positively charged peptide-presenting T7 has a high number of intra-tissue binding sites in the TME (~4 µM) that enables almost 100% retention in the tumor tissue for up to 24 h. These results, coupled with transport studies of systematically mutated T7, show that electrostatic interactions can be responsible for uptake and retention of the positively charged peptide-presenting T7 within the net negatively charged TME. STATEMENT OF SIGNIFICANCE: The TME selectively hinders the transport of drugs and drug delivery systems due to their size, shape, and intermolecular interactions. Typically, the focus in drug delivery has been to develop delivery systems smaller than the pore size of the tumor ECM and/or develop inert surface coatings that have negligible interactions with the tumor ECM for diffusive transport. While there is an association of the surface charge of carriers with their transport through the tumor ECM, the mechanism of charge-driven transport is poorly understood. In this work, we elucidate the mechanism and find that interestingly, particles with a weakly positive surface charge interact with the net negatively charged tumor ECM to significantly improve their uptake, penetration, and retention in tumor tissue.
Collapse
|
42
|
Liu Y, Yang G, Jin S, Xu L, Zhao CX. Development of High-Drug-Loading Nanoparticles. Chempluschem 2020; 85:2143-2157. [PMID: 32864902 DOI: 10.1002/cplu.202000496] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Formulating drugs into nanoparticles offers many attractive advantages over free drugs including improved bioavailability, minimized toxic side effects, enhanced drug delivery, feasibility of incorporating other functions such as controlled release, imaging agents for imaging, targeting delivery, and loading more than one drug for combination therapies. One of the key parameters is drug loading, which is defined as the mass ratio of drug to drug-loaded nanoparticles. Currently, most nanoparticle systems have relatively low drug loading (<10 wt%), and developing methods to increase drug loading remains a challenge. This Minireview presents an overview of recent research on developing nanoparticles with high drug loading (>10 wt%) from the perspective of synthesis strategies, including post-loading, co-loading, and pre-loading. Based on these three different strategies, various nanoparticle systems with different materials and drugs are summarized and discussed in terms of their synthesis methods, drug loadings, encapsulation efficiencies, release profiles, stabilities, and their applications in drug delivery. The advantages and disadvantages of these strategies are presented with an objective of providing useful design rules for future development of high-drug-loading nanoparticles.
Collapse
Affiliation(s)
- Yun Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Song Jin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Letao Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| |
Collapse
|
43
|
Advances in Gold Nanoparticle-Based Combined Cancer Therapy. NANOMATERIALS 2020; 10:nano10091671. [PMID: 32858957 PMCID: PMC7557687 DOI: 10.3390/nano10091671] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
According to the global cancer observatory (GLOBOCAN), there are approximately 18 million new cancer cases per year worldwide. Cancer therapies are largely limited to surgery, radiotherapy, and chemotherapy. In radiotherapy and chemotherapy, the maximum tolerated dose is presently being used to treat cancer patients. The integrated development of innovative nanoparticle (NP) based approaches will be a key to address one of the main issues in both radiotherapy and chemotherapy: normal tissue toxicity. Among other inorganic NP systems, gold nanoparticle (GNP) based systems offer the means to further improve chemotherapy through controlled delivery of chemotherapeutics, while local radiotherapy dose can be enhanced by targeting the GNPs to the tumor. There have been over 20 nanotechnology-based therapeutic products approved for clinical use in the past two decades. Hence, the goal of this review is to understand what we have achieved so far and what else we can do to accelerate clinical use of GNP-based therapeutic platforms to minimize normal tissue toxicity while increasing the efficacy of the treatment. Nanomedicine will revolutionize future cancer treatment options and our ultimate goal should be to develop treatments that have minimum side effects, for improving the quality of life of all cancer patients.
Collapse
|
44
|
Mao S, Pang Y, Liu T, Shao Y, He J, Yang H, Mao Y, Sun W. Bioprinting of in vitro tumor models for personalized cancer treatment: a review. Biofabrication 2020; 12:042001. [PMID: 32470967 DOI: 10.1088/1758-5090/ab97c0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Studying biological characteristics of tumors and evaluating the treatment effects require appropriate in vitro tumor models. However, the occurrence, progression, and migration of tumors involve spatiotemporal changes, cell-microenvironment and cell-cell interactions, and signal transmission in cells, which makes the construction of in vitro tumor models extremely challenging. In the past few years, advances in biomaterials and tissue engineering methods, especially development of the bioprinting technology, have paved the way for innovative platform technologies for in vitro cancer research. Bioprinting can accurately control the distribution of cells, active molecules, and biomaterials. Furthermore, this technology recapitulates the key characteristics of the tumor microenvironment and constructs in vitro tumor models with bionic structures and physiological systems. These models can be used as robust platforms to study tumor initiation, interaction with the microenvironment, angiogenesis, motility and invasion, as well as intra- and extravasation. Bioprinted tumor models can also be used for high-throughput drug screening and validation and provide the possibility for personalized cancer treatment research. This review describes the basic characteristics of the tumor and its microenvironment and focuses on the importance and relevance of bioprinting technology in the construction of tumor models. Research progress in the bioprinting of monocellular, multicellular, and personalized tumor models is discussed, and comprehensive application of bioprinting in preclinical drug screening and innovative therapy is reviewed. Finally, we offer our perspective on the shortcomings of the existing models and explore new technologies to outline the direction of future development and application prospects of next-generation tumor models.
Collapse
Affiliation(s)
- Shuangshuang Mao
- Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, Beijing, People's Republic of China. Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, People's Republic of China. 'Biomanufacturing and Engineering Living Systems' 111 -Innovation International Talents Base, Beijing, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Imaging using radiolabelled targeted proteins: radioimmunodetection and beyond. EJNMMI Radiopharm Chem 2020; 5:16. [PMID: 32577943 PMCID: PMC7311618 DOI: 10.1186/s41181-020-00094-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/14/2020] [Indexed: 12/18/2022] Open
Abstract
The use of radiolabelled antibodies was proposed in 1970s for staging of malignant tumours. Intensive research established chemistry for radiolabelling of proteins and understanding of factors determining biodistribution and targeting properties. The use of radioimmunodetection for staging of cancer was not established as common practice due to approval and widespread use of [18F]-FDG, which provided a more general diagnostic use than antibodies or their fragments. Expanded application of antibody-based therapeutics renewed the interest in radiolabelled antibodies. RadioimmunoPET emerged as a powerful tool for evaluation of pharmacokinetics of and target engagement by biotherapeutics. In addition to monoclonal antibodies, new radiolabelled engineered proteins have recently appeared, offering high-contrast imaging of expression of therapeutic molecular targets in tumours shortly after injection. This creates preconditions for noninvasive determination of a target expression level and stratification of patients for targeted therapies. Radiolabelled proteins hold great promise to play an important role in development and implementation of personalised targeted treatment of malignant tumours. This article provides an overview of biodistribution and tumour-seeking features of major classes of targeting proteins currently utilized for molecular imaging. Such information might be useful for researchers entering the field of the protein-based radionuclide molecular imaging.
Collapse
|
46
|
Mohammadabadi A, Huynh RN, Wadajkar AS, Lapidus RG, Kim AJ, Raub CB, Frenkel V. Pulsed focused ultrasound lowers interstitial fluid pressure and increases nanoparticle delivery and penetration in head and neck squamous cell carcinoma xenograft tumors. Phys Med Biol 2020; 65:125017. [PMID: 32460260 DOI: 10.1088/1361-6560/ab9705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanocarriers offer a promising approach to significantly improve therapeutic delivery to solid tumors as well as limit the side effects associated with anti-cancer agents. However, their relatively large size can negatively affect their ability to efficiently penetrate into more interior tumor regions, ultimately reducing therapeutic efficacy. Poor penetration of large agents such as nanocarriers is attributed to factors in the tumor microenvironment such as elevated interstitial fluid pressure (IFP) and fibrillar collagen in the extracellular matrix. Our previous studies reported that pretreatment of solid tumor xenografts with nondestructive pulsed focused ultrasound (pFUS) can improve the delivery and subsequent therapy of a variety of therapeutic formulations in different tumor models, where the results were associated with expanded extracellular spaces (ECS), an increase in hydraulic conductivity, and decrease in tissue stiffness. Here, we demonstrate the inverse relationship between IFP and the penetration of systemically administered nanoparticle (NP) probes, where IFP increased from the tumor periphery to their center. Furthermore, we show that pretreatment with pFUS can safely reduce IFP and improve NP delivery; especially into the center of the tumors. These results coincide with effects generated in the fibrillar collagen network microstructure in the ECS as determined by quantitative polarized light microscopy. Whole tumor and histomorphometric analysis, however, did not show significant differences in collagen area fraction or collagen feature solidity, as well as tumor cross-sectional area and aspect ratio, as a result of the treatments. We present a biophysical model connecting the experimental results, where pFUS-mediated cytoarchitectural changes are associated with improved redistribution of the interstitial fluid and lower IFP. The resulting improvement in NP delivery supports our previous therapeutic studies and may have implications for clinical applications to improve therapeutic outcomes in cancer therapy.
Collapse
Affiliation(s)
- Ali Mohammadabadi
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States of America. Department of Mechanical Engineering, University of Maryland, Baltimore County, Catonsville, MD, United States of America
| | | | | | | | | | | | | |
Collapse
|
47
|
Yang G, Liu Y, Jin S, Zhao C. Development of Core‐Shell Nanoparticle Drug Delivery Systems Based on Biomimetic Mineralization. Chembiochem 2020; 21:2871-2879. [DOI: 10.1002/cbic.202000105] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 04/28/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia, Queensland 4072 Australia
| | - Yun Liu
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia, Queensland 4072 Australia
| | - Song Jin
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia, Queensland 4072 Australia
| | - Chun‐Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia, Queensland 4072 Australia
| |
Collapse
|
48
|
Sindhwani S, Syed AM, Ngai J, Kingston BR, Maiorino L, Rothschild J, MacMillan P, Zhang Y, Rajesh NU, Hoang T, Wu JLY, Wilhelm S, Zilman A, Gadde S, Sulaiman A, Ouyang B, Lin Z, Wang L, Egeblad M, Chan WCW. The entry of nanoparticles into solid tumours. NATURE MATERIALS 2020; 19:566-575. [PMID: 31932672 DOI: 10.1038/s41563-019-0566-2] [Citation(s) in RCA: 1038] [Impact Index Per Article: 207.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 11/15/2019] [Indexed: 05/20/2023]
Abstract
The concept of nanoparticle transport through gaps between endothelial cells (inter-endothelial gaps) in the tumour blood vessel is a central paradigm in cancer nanomedicine. The size of these gaps was found to be up to 2,000 nm. This justified the development of nanoparticles to treat solid tumours as their size is small enough to extravasate and access the tumour microenvironment. Here we show that these inter-endothelial gaps are not responsible for the transport of nanoparticles into solid tumours. Instead, we found that up to 97% of nanoparticles enter tumours using an active process through endothelial cells. This result is derived from analysis of four different mouse models, three different types of human tumours, mathematical simulation and modelling, and two different types of imaging techniques. These results challenge our current rationale for developing cancer nanomedicine and suggest that understanding these active pathways will unlock strategies to enhance tumour accumulation.
Collapse
Affiliation(s)
- Shrey Sindhwani
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Abdullah Muhammad Syed
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jessica Ngai
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin R Kingston
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Laura Maiorino
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- Watson School of Biological Sciences, Cold Spring Harbor, NY, USA
| | - Jeremy Rothschild
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Presley MacMillan
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yuwei Zhang
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Netra Unni Rajesh
- Division of Engineering Science, University of Toronto, Toronto, Ontario, Canada
| | - Tran Hoang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jamie L Y Wu
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Anton Zilman
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Suresh Gadde
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Andrew Sulaiman
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Ben Ouyang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Zachary Lin
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Warren C W Chan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
- Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Materials Science and Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Ahmad T, Sarwar R, Iqbal A, Bashir U, Farooq U, Halim SA, Khan A, Al-Harrasi A. Recent advances in combinatorial cancer therapy via multifunctionalized gold nanoparticles. Nanomedicine (Lond) 2020; 15:1221-1237. [DOI: 10.2217/nnm-2020-0051] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The diverse behavior of nanogold in the therapeutic field is related to its unique size and shape. Nanogold offers improvements in modern diagnostic and therapeutic implications, increases disease specificity and targeted drug delivery, and is relatively economical compared with other chemotherapeutic protocols. The diagnosis of cancer and photothermal therapy improve drastically with the implementation of nanotechnology. Different types of nanoparticles, that is, gold silica nanoshells, nanorods and nanospheres of diverse shapes and geometries, are used widely in the photothermal therapy of cancerous cells and nodules. Numerous reviews have been published on the therapeutic applications of gold nanoparticles, but studies on combinatorial applications of nanogold in cancer therapy are limited. This review focuses on the combinatorial cancer therapy using optical properties of nanogold with different shapes and geometries, and their therapeutic applications in cancer diagnosis, photothermal therapy, cancer imaging and targeted drug delivery.
Collapse
Affiliation(s)
- Touqeer Ahmad
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Rizwana Sarwar
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Ayesha Iqbal
- Division of Pharmacy Practice & Policy, School of pharmacy, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Uzma Bashir
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Umar Farooq
- Department of Chemistry, COMSATS University Islamabad Abbottabad Campus, Abbottabad, Pakistan
| | - Sobia Ahsan Halim
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Ajmal Khan
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Center, University of Nizwa, PO Box 33, Birkat Al Mauz, Nizwa, 616, Sultanate of Oman
| |
Collapse
|
50
|
Shirai H, Tsukada K. Bacterial proteolytic activity improves drug delivery in tumors in a size, pharmacokinetic, and binding affinity dependent manner - A mechanistic understanding. J Control Release 2020; 321:348-362. [PMID: 32061790 DOI: 10.1016/j.jconrel.2020.02.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/15/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023]
Abstract
Motile bacteria are able to penetrate in the distal areas of blood vessel, which makes bacteria attractive to researchers as a drug delivery vehicle carrying anti-cancer drugs to tumors. Not only therapeutic bacteria show wide anti-tumor effect but also the combination of therapeutic bacteria and conventional chemotherapy leads to dramatically large synergetic effect. We provide a mechanistic understanding of enhanced drug delivery in tumors by co-administration of chemotherapeutic agents and therapeutic bacteria. In this work, simultaneous delivery of C. novyi-NT and chemotherapeutic agents in tumors is mathematically modeled. Simulated doxorubicin concentration in tumors after Doxil administration with or without bacteria agreed reasonably well with experimental literature. Simulated doxorubicin concentration in tumors by the combination of Doxil and C. novyi-NT is over twice higher than that of Doxil alone. This enhanced doxorubicin concentration in tumors is due to the degradation of extracellular matrix of collagen by bacterial proteolytic activity, which increases hydraulic conductivity of interstitium, reduces interstitial fluid pressure, and thus increases convection through vessel walls. Additionally, it alleviates solid stress, which decompresses blood vessels, and thus increases vessel density. On the other hand, simulated doxorubicin concentration in tumors for non-liposomal free-doxorubicin is not enhanced by C. novyi-NT because vascular permeability of free-doxorubicin is larger than Doxil, and thus increased but relatively small convection across vessel walls is offset by the efflux due to increased interstitial flow. A strategy to further enhance this combination therapy is discussed along with sensitivity analysis.
Collapse
Affiliation(s)
- Hiroaki Shirai
- Graduates School of Science and Technology, Keio University, 3-14-1 Hiyoshi Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan.
| | - Kosuke Tsukada
- Graduates School of Science and Technology, Keio University, 3-14-1 Hiyoshi Kohoku-ku, Yokohama-shi, Kanagawa 223-8522, Japan
| |
Collapse
|