1
|
Liang B, Xu Y, Yu N, Yang Z, Wilson M, Xu D, Shams RA, Wang L, Lui CHJ, Yan R, Liu M. Enhanced Plasmonic Trapping and Fluorescent Emission of Nitrogen-Vacancy Nanodiamonds Using a High-Efficiency Nanofocusing Device. NANO LETTERS 2024; 24:11661-11668. [PMID: 39250914 PMCID: PMC11421074 DOI: 10.1021/acs.nanolett.4c03163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024]
Abstract
Fluorescent nanodiamonds (FNDs) with nitrogen-vacancy centers are pivotal for advancing quantum photonics and imaging through deterministic quantum state manipulation. However, deterministic integration of quantum emitters into photonic devices remains a challenge due to the need for high coupling efficiency and Purcell enhancement. We report a deterministic FND-integrated nanofocusing device achieved by assembling FNDs at a plasmonic waveguide tip through plasmonic-enhanced optical trapping. This technique not only increases the emission rate by 58.6 times compared to isolated FNDs but also preferentially directs radiation into the waveguide at a rate 5.3 times higher than that into free space, achieving an exceptional figure-of-merit of ∼3000 for efficient energy transfer. Our findings represent a significant step toward deterministic integration in quantum imaging and communication, opening new avenues for quantum technology advancements.
Collapse
Affiliation(s)
- Boqun Liang
- Materials
Science and Engineering Program, University
of California−Riverside, Riverside, California 92521, United States
| | - Yaodong Xu
- Materials
Science and Engineering Program, University
of California−Riverside, Riverside, California 92521, United States
| | - Ning Yu
- Department
of Chemical and Environmental Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Zhaoxi Yang
- Department
of Chemical and Environmental Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Matthew Wilson
- Department
of Physics and Astronomy, University of
California−Riverside, Riverside, California 92521, United States
| | - Da Xu
- Department
of Electrical and Computer Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Rifat Ara Shams
- Department
of Electrical and Computer Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Longjian Wang
- Department
of Electrical and Computer Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Chun Hung Joshua Lui
- Department
of Physics and Astronomy, University of
California−Riverside, Riverside, California 92521, United States
| | - Ruoxue Yan
- Department
of Chemical and Environmental Engineering, University of California−Riverside, Riverside, California 92521, United States
- Department
of Electrical and Computer Engineering, University of California−Riverside, Riverside, California 92521, United States
| | - Ming Liu
- Materials
Science and Engineering Program, University
of California−Riverside, Riverside, California 92521, United States
- Department
of Electrical and Computer Engineering, University of California−Riverside, Riverside, California 92521, United States
| |
Collapse
|
2
|
Kohaar I, Hodges NA, Srivastava S. Biomarkers in Cancer Screening: Promises and Challenges in Cancer Early Detection. Hematol Oncol Clin North Am 2024; 38:869-888. [PMID: 38782647 PMCID: PMC11222039 DOI: 10.1016/j.hoc.2024.04.004] [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] [Indexed: 05/25/2024]
Abstract
Cancer continues to be one the leading causes of death worldwide, primarily due to the late detection of the disease. Cancers detected at early stages may enable more effective intervention of the disease. However, most cancers lack well-established screening procedures except for cancers with an established early asymptomatic phase and clinically validated screening tests. There is a critical need to identify and develop assays/tools in conjunction with imaging approaches for precise screening and detection of the aggressive disease at an early stage. New developments in molecular cancer screening and early detection include germline testing, synthetic biomarkers, and liquid biopsy approaches.
Collapse
Affiliation(s)
- Indu Kohaar
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA
| | - Nicholas A Hodges
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, NIH, 9609 Medical Center Drive, NCI Shady Grove Building, Rockville, MD 20850, USA.
| |
Collapse
|
3
|
Fan D, Cao Y, Cao M, Wang Y, Cao Y, Gong T. Nanomedicine in cancer therapy. Signal Transduct Target Ther 2023; 8:293. [PMID: 37544972 PMCID: PMC10404590 DOI: 10.1038/s41392-023-01536-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/08/2023] Open
Abstract
Cancer remains a highly lethal disease in the world. Currently, either conventional cancer therapies or modern immunotherapies are non-tumor-targeted therapeutic approaches that cannot accurately distinguish malignant cells from healthy ones, giving rise to multiple undesired side effects. Recent advances in nanotechnology, accompanied by our growing understanding of cancer biology and nano-bio interactions, have led to the development of a series of nanocarriers, which aim to improve the therapeutic efficacy while reducing off-target toxicity of the encapsulated anticancer agents through tumor tissue-, cell-, or organelle-specific targeting. However, the vast majority of nanocarriers do not possess hierarchical targeting capability, and their therapeutic indices are often compromised by either poor tumor accumulation, inefficient cellular internalization, or inaccurate subcellular localization. This Review outlines current and prospective strategies in the design of tumor tissue-, cell-, and organelle-targeted cancer nanomedicines, and highlights the latest progress in hierarchical targeting technologies that can dynamically integrate these three different stages of static tumor targeting to maximize therapeutic outcomes. Finally, we briefly discuss the current challenges and future opportunities for the clinical translation of cancer nanomedicines.
Collapse
Affiliation(s)
- Dahua Fan
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China.
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meiqun Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Yajun Wang
- Shunde Women and Children's Hospital, Guangdong Medical University, Foshan, 528300, China
| | | | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
4
|
Cheng Y, Sun C, Chang Y, Wu J, Zhang Z, Liu Y, Ge S, Li Z, Li X, Sun L, Zang D. Photoelectrochemical biosensor based on SiW 12@CdS quantum dots for the highly sensitive detection of HPV 16 DNA. Front Bioeng Biotechnol 2023; 11:1193052. [PMID: 37388766 PMCID: PMC10303914 DOI: 10.3389/fbioe.2023.1193052] [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: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
A highly sensitive biosensor for detecting HPV 16 DNA was prepared based on Keggin-type polyoxometalate (SiW12)-grafted CdS quantum dots (SiW12@CdS QDs) and colloidal gold nanoparticles (Au NPs), which exhibited remarkable selectivity and sensitivity upon target DNA detection because of its excellent photoelectrochemical (PEC) response. Here, an enhanced photoelectronic response ability was achieved with the strong association of SiW12@CdS QDs by polyoxometalate modification, which was developed through a convenient hydrothermal process. Furthermore, on Au NP-modified indium tin oxide slides, a multiple-site tripodal DNA walker sensing platform coupled with T7 exonuclease was successfully fabricated with SiW12@CdS QDs/NP DNA as a probe for detecting HPV 16 DNA. Due to the remarkable conductivity of Au NPs, the photosensitivity of the as-prepared biosensor was improved in an I3-/I- solution and avoided the use of other regents toxic to living organisms. Finally, under optimized conditions, the as-prepared biosensor protocol demonstrated wide linear ranges (15-130 nM), with a limit of detection of 0.8 nM and high selectivity, stability, and reproducibility. Moreover, the proposed PEC biosensor platform offers a reliable pathway for detecting other biological molecules with nano-functional materials.
Collapse
Affiliation(s)
- Yao Cheng
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chaoyue Sun
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, China
| | - Yuhua Chang
- Shandong Provincial Maternal and Child Healthcare Hospital, Jinan, China
| | - Jiayin Wu
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhihao Zhang
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yunqing Liu
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, China
| | - Shenguang Ge
- Institute for Advanced Interdisciplinary Research (iAIR), School of Chemistry and Chemical Engineering, University of Jinan, Jinan, China
| | - Zhao Li
- Suzhou KunTao Intelligent Manufacturing Technology Co., Ltd., Suzhou, China
| | - Xiao Li
- NMPA Key Laboratory for Quality Evaluation of Medical Materials and Biological Protective Devices, Jinan, China
- Shandong Institute of Medical Device and Pharmaceutical Packaging Inspection, Jinan, China
| | - Liang Sun
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Dejin Zang
- National Key Laboratory of Advanced Drug Delivery and Release System, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, School of Pharmacy and Pharmaceutical Sciences, Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
5
|
Hsu JC, Tang Z, Eremina OE, Sofias AM, Lammers T, Lovell JF, Zavaleta C, Cai W, Cormode DP. Nanomaterial-based contrast agents. NATURE REVIEWS. METHODS PRIMERS 2023; 3:30. [PMID: 38130699 PMCID: PMC10732545 DOI: 10.1038/s43586-023-00211-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/20/2023] [Indexed: 12/23/2023]
Abstract
Medical imaging, which empowers the detection of physiological and pathological processes within living subjects, has a vital role in both preclinical and clinical diagnostics. Contrast agents are often needed to accompany anatomical data with functional information or to provide phenotyping of the disease in question. Many newly emerging contrast agents are based on nanomaterials as their high payloads, unique physicochemical properties, improved sensitivity and multimodality capacity are highly desired for many advanced forms of bioimaging techniques and applications. Here, we review the developments in the field of nanomaterial-based contrast agents. We outline important nanomaterial design considerations and discuss the effect on their physicochemical attributes, contrast properties and biological behaviour. We also describe commonly used approaches for formulating, functionalizing and characterizing these nanomaterials. Key applications are highlighted by categorizing nanomaterials on the basis of their X-ray, magnetic, nuclear, optical and/or photoacoustic contrast properties. Finally, we offer our perspectives on current challenges and emerging research topics as well as expectations for future advancements in the field.
Collapse
Affiliation(s)
- Jessica C. Hsu
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Zhongmin Tang
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Olga E. Eremina
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - David P. Cormode
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
6
|
Assessment of the Pharmacokinetics, Disposition, and Duration of Action of the Tumour-Targeting Peptide CEND-1. Int J Mol Sci 2023; 24:ijms24065700. [PMID: 36982773 PMCID: PMC10053770 DOI: 10.3390/ijms24065700] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
CEND-1 (iRGD) is a bifunctional cyclic peptide that can modulate the solid tumour microenvironment, enhancing the delivery and therapeutic index of co-administered anti-cancer agents. This study explored CEND-1’s pharmacokinetic (PK) properties pre-clinically and clinically, and assessed CEND-1 distribution, tumour selectivity and duration of action in pre-clinical tumour models. Its PK properties were assessed after intravenous infusion of CEND-1 at various doses in animals (mice, rats, dogs and monkeys) and patients with metastatic pancreatic cancer. To assess tissue disposition, [3H]-CEND-1 radioligand was administered intravenously to mice bearing orthotopic 4T1 mammary carcinoma, followed by tissue measurement using quantitative whole-body autoradiography or quantitative radioactivity analysis. The duration of the tumour-penetrating effect of CEND-1 was evaluated by assessing tumour accumulation of Evans blue and gadolinium-based contrast agents in hepatocellular carcinoma (HCC) mouse models. The plasma half-life was approximately 25 min in mice and 2 h in patients following intravenous administration of CEND-1. [3H]-CEND-1 localised to the tumour and several healthy tissues shortly after administration but was cleared from most healthy tissues by 3 h. Despite the rapid systemic clearance, tumours retained significant [3H]-CEND-1 several hours post-administration. In mice with HCC, the tumour penetration activity remained elevated for at least 24 h after the injection of a single dose of CEND-1. These results indicate a favourable in vivo PK profile of CEND-1 and a specific and sustained tumour homing and tumour penetrability. Taken together, these data suggest that even single injections of CEND-1 may elicit long-lasting tumour PK improvements for co-administered anti-cancer agents.
Collapse
|
7
|
Li S, Wei J, Yao Q, Song X, Xie J, Yang H. Emerging ultrasmall luminescent nanoprobes for in vivo bioimaging. Chem Soc Rev 2023; 52:1672-1696. [PMID: 36779305 DOI: 10.1039/d2cs00497f] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Photoluminescence (PL) imaging has become a fundamental tool in disease diagnosis, therapeutic evaluation, and surgical navigation applications. However, it remains a big challenge to engineer nanoprobes for high-efficiency in vivo imaging and clinical translation. Recent years have witnessed increasing research efforts devoted into engineering sub-10 nm ultrasmall nanoprobes for in vivo PL imaging, which offer the advantages of efficient body clearance, desired clinical translation potential, and high imaging signal-to-noise ratio. In this review, we present a comprehensive summary and contrastive discussion of emerging ultrasmall luminescent nanoprobes towards in vivo PL bioimaging of diseases. We first summarize size-dependent nano-bio interactions and imaging features, illustrating the unique attributes and advantages/disadvantages of ultrasmall nanoprobes differentiating them from molecular and large-sized probes. We also discuss general design methodologies and PL properties of emerging ultrasmall luminescent nanoprobes, which are established based on quantum dots, metal nanoclusters, lanthanide-doped nanoparticles, and silicon nanoparticles. Then, recent advances of ultrasmall luminescent nanoprobes are highlighted by surveying their latest in vivo PL imaging applications. Finally, we discuss existing challenges in this exciting field and propose some strategies to improve in vivo PL bioimaging and further propel their clinical applications.
Collapse
Affiliation(s)
- Shihua Li
- Qingyuan Innovation Laboratory, 1# Xueyuan Road, Quanzhou, Fujian 362801, China.,MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China.
| | - Jing Wei
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China. .,Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Qiaofeng Yao
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore. .,Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, Fujian 350207, China
| | - Xiaorong Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China. .,Fujian Science &Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian 350108, China
| | - Jianping Xie
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore. .,Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Binhai New City, Fuzhou, Fujian 350207, China
| | - Huanghao Yang
- Qingyuan Innovation Laboratory, 1# Xueyuan Road, Quanzhou, Fujian 362801, China.,MOE Key Laboratory for Analytical Science of Food Safety and Biology, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350116, China. .,Fujian Science &Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou, Fujian 350108, China
| |
Collapse
|
8
|
Han Y, Yi H, Wang Y, Li Z, Chu X, Jiang JH. Ultrathin Zinc Selenide Nanoplatelets Boosting Photoacoustic Imaging of In Situ Copper Exchange in Alzheimer's Disease Mice. ACS NANO 2022; 16:19053-19066. [PMID: 36349982 DOI: 10.1021/acsnano.2c08094] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The critical role of transition metal dyshomeostasis in Alzheimer's disease (AD) pathology poses demands of in vivo imaging for brain copper levels. Nanostructured probes afford prolonged retention time, increased accumulation, and enhanced photostability; however, their development for activatable photoacoustic (PA) imaging remains largely unexplored. We develop a principle of concept for activable PA imaging using in situ cation exchange of ultrathin zinc selenide (ZnSe) nanoplatelets for monitoring brain copper levels in AD mice. We start from quantitative modeling of optical absorption, time-resolved temperature field, and thermal expansion of copper selenide (CuSe) nanocrystals of different morphologies and reveal that ultrathin nanoplatelets afford substantial enhancement of near-infrared (NIR) absorption and PA pressures as compared to nanodots and nanoparticles. By tethering with a blood-brain barrier (BBB)-targeting peptide ligand, the ultrathin ZnSe nanoplatelet probe efficiently transports across the BBB and rapidly exchanges with endogenous copper ions, boosting activatable PA imaging of brain copper levels. We also demonstrate that the efficient exchange of ZnSe nanoplatelets with copper ions can reduce oxidative stress of neurons and protect neuronal cells from apoptosis. The nanoplatelet probe provides a paradigm for activatable PA imaging of brain copper levels, highlighting its potential for pathophysiologic study of AD.
Collapse
Affiliation(s)
- Yajing Han
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Haoyu Yi
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Yujie Wang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Zheng Li
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Xia Chu
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| | - Jian-Hui Jiang
- State Key Laboratory of Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, People's Republic of China
| |
Collapse
|
9
|
Dean A, Gill S, McGregor M, Broadbridge V, Järveläinen HA, Price T. Dual αV-integrin and neuropilin-1 targeting peptide CEND-1 plus nab-paclitaxel and gemcitabine for the treatment of metastatic pancreatic ductal adenocarcinoma: a first-in-human, open-label, multicentre, phase 1 study. Lancet Gastroenterol Hepatol 2022; 7:943-951. [PMID: 35803294 DOI: 10.1016/s2468-1253(22)00167-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND CEND-1 is a novel cyclic peptide that targets αV integrins and neuropilin-1 and enhances tumour delivery of co-administered anticancer drugs. We investigated the safety, tolerability, and biological activity of CEND-1 in patients with metastatic pancreatic ductal adenocarcinoma in combination with nab-paclitaxel and gemcitabine. METHODS This open-label, multicentre, phase 1 study, conducted at three hospitals in Australia, enrolled participants aged 18 years or older with histologically confirmed metastatic pancreatic ductal adenocarcinoma who had one or more lesions measurable on MRI or CT, an Eastern Cooperative Oncology Group performance status score of 0 or 1, and a life expectancy of at least 3 months. Exclusion criteria included previous chemotherapy and brain metastases or other malignancy (unless receiving curative intent). There was no randomisation or masking. CEND-1 monotherapy was given as an intravenous fluid bolus on day 1 of a run-in phase of 7 days (0·2-3·2 mg/kg) followed by CEND-1 plus intravenous gemcitabine (1000 mg/m2) and nab-paclitaxel (125 mg/m2) on days 1, 8, and 15 of 28-day treatment cycles until disease progression. The primary safety endpoints were incidence, severity, and duration of treatment-emergent and treatment-related adverse events; overall survival; and clinical laboratory results, which were all assessed in the safety population. This study is registered with ClinicalTrials.gov, NCT03517176, and the Australian New Zealand Clinical Trials Registry, ACTRN12618000804280. FINDINGS Between Aug 13, 2018, and Nov 30, 2019, 31 patients were enrolled (eight in the dose-escalation phase [cohort 1a] and 23 in the expansion phase [cohort 1b]). Two patients were excluded from the efficacy population. No CEND-1 dose-limiting toxicities were observed in the safety population (n=31). The most common grade 3 or 4 events were neutropenia (17 [55%] patients), anaemia (eight [26%]), leukopenia (five [16%]), and pulmonary embolism (four [13%]). Serious adverse events occurred in 22 (71%) patients, mostly related to disease progression. Ten deaths occurred during the study due to progression of metastatic pancreatic cancer (n=9) and a left middle cerebral artery stroke (n=1). In the efficacy population (n=29), 17 (59%) patients had an objective response, including one complete response and 16 partial responses. After a median follow-up of 26 months (IQR 24-30), median overall survival was 13·2 months (95% CI 9·7-22·5). INTERPRETATION CEND-1 with nab-paclitaxel and gemcitabine has an acceptable safety profile, with no dose-limiting toxicities and encouraging activity. Adverse events were generally consistent with those seen with nab-paclitaxel and gemcitabine. Further randomised trials to determine the efficacy of CEND-1 are warranted. FUNDING DrugCendR Australia Pty.
Collapse
Affiliation(s)
- Andrew Dean
- Bendat Family Comprehensive Cancer Centre, St John of God Subiaco Hospital, Subiaco, WA, Australia.
| | - Sanjeev Gill
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
| | - Mark McGregor
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Vy Broadbridge
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Harri A Järveläinen
- DrugCendR Australia Pty, Collingwood, VIC, Australia; Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Timothy Price
- Department of Medical Oncology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| |
Collapse
|
10
|
Hesemans E, Buttiens K, Manshian BB, Soenen SJ. The Role of Optical Imaging in Translational Nanomedicine. J Funct Biomater 2022; 13:137. [PMID: 36135572 PMCID: PMC9502568 DOI: 10.3390/jfb13030137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Nanomedicines have been a major research focus in the past two decades and are increasingly emerging in a broad range of clinical applications. However, a proper understanding of their biodistribution is required to further progress the field of nanomedicine. For this, imaging methods to monitor the delivery and therapeutic efficacy of nanoparticles are urgently needed. At present, optical imaging is the most common method used to study the biodistribution of nanomaterials, where the unique properties of nanomaterials and advances in optical imaging can jointly result in novel methods for optimal monitoring of nanomaterials in preclinical animal models. This review article aims to give an introduction to nanomedicines and their translational impact to highlight the potential of optical imaging to study the biodistribution of nanoparticles and to monitor the delivery and therapeutic efficacy at the preclinical level. After introducing both domains, the review focuses on different techniques that can be used to overcome some intrinsic limitations of optical imaging and how this can specifically benefit nanoparticle studies. Finally, we point out some important key features of nanoparticles that currently hinder their full potential in the clinic and how the advances in optical imaging can help to provide us with the information needed to further boost the clinical translation and expand the field of nanomedicines.
Collapse
Affiliation(s)
- Evelien Hesemans
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Kiana Buttiens
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
- Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
- Leuven Cancer Institute, 3000 Leuven, Belgium
| |
Collapse
|
11
|
Sun Z, Wang Z, Wang T, Wang J, Zhang H, Li Z, Wang S, Sheng F, Yu J, Hou Y. Biodegradable MnO-Based Nanoparticles with Engineering Surface for Tumor Therapy: Simultaneous Fenton-Like Ion Delivery and Immune Activation. ACS NANO 2022; 16:11862-11875. [PMID: 35925671 DOI: 10.1021/acsnano.2c00969] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immune checkpoint inhibitors have achieved significant clinical success but are still suffering from inadequate immune activation. It is worth noting that manganese as a nutritional inorganic trace element is closely associated with immune activation to fight against tumor growth and metastasis via the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. Herein, we designed hollow mesoporous silica-coated MnO nanoparticles (NPs), followed by conjugation of tumor homing peptide iRGD (CRGDKGPD). The obtained NPs (MnO@mSiO2-iRGD NPs) were applied to magnetic resonance imaging (MRI)-guided tumor immune-chemodynamic combination therapy, in which MnO NPs can be harnessed for cGAS-STING pathway-activated immunotherapy, Fenton-like reaction-induced reactive oxygen species upregulation, and T1-weighted MRI. The rough surface and large cavities of the mSiO2 shell promote cellular uptake and MnO NPs delivery. Meanwhile, it was found that MnO@mSiO2-iRGD NPs would dissociate under an acid environment, resulting in tumor specificity of MRI and exogenous Mn2+ release. Our results revealed that these pH-responsive biodegradable MnO@mSiO2-iRGD NPs synergized with α-PD-1 (PD-1 = programmed cell death-1) blocking antibody to highly elicit cytotoxic T lymphocyte infiltration and restrict melanoma progression and metastasis, which were envisioned as a promising candidate for tumor theranostics.
Collapse
Affiliation(s)
- Zhaoli Sun
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, School of Life Sciences, Peking University, Beijing 100871, China
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Zhiyi Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Tao Wang
- Electron Microscopy Laboratory, School of Physics, Peking University, Beijing 100871, China
| | - Jingjing Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Hongtao Zhang
- Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Ziyuan Li
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Shuren Wang
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Fugeng Sheng
- Fifth Medical Center, Chinese PLA General Hospital, Beijing 100039, China
| | - Jing Yu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yanglong Hou
- Beijing Key Laboratory for Magnetoelectric Materials and Devices, School of Materials Science and Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Abstract
The term "molecular ZIP (or area) codes" refers to an originally hypothetical system of cell adhesion molecules that would control cell trafficking in the body. Subsequent discovery of the integrins, cadherins, and other cell adhesion molecules confirmed this hypothesis. The recognition system encompassing integrins and their ligands came particularly close to fulfilling the original ZIP code hypothesis, as multiple integrins with closely related specificities mediate cell adhesion by binding to an RGD or related sequence in various extracellular matrix proteins. Diseased tissues have their own molecular addresses that, although not necessarily involved in cell trafficking, can be made use of in targeted drug delivery. This article discusses the molecular basis of ZIP codes and the extensive effort under way to harness them for drug delivery purposes.
Collapse
|
13
|
Paithankar JG, Kushalan S, S N, Hegde S, Kini S, Sharma A. Systematic toxicity assessment of CdTe quantum dots in Drosophila melanogaster. CHEMOSPHERE 2022; 295:133836. [PMID: 35120950 DOI: 10.1016/j.chemosphere.2022.133836] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/31/2022] [Accepted: 01/31/2022] [Indexed: 06/14/2023]
Abstract
The risk assessment of cadmium (Cd)-based quantum dots (QDs) used for biomedical nanotechnology applications has stern toxicity concerns. Despite cytotoxicity studies of cadmium telluride (CdTe) QDs, the systematic in vivo study focusing on its organismal effects are more relevant to public health. Therefore, the present study aims to investigate the effect of chemically synthesized 3-mercapto propionic acid-functionalized CdTe QDs on organisms' survival, development, reproduction, and behaviour using Drosophila melanogaster as a model. The sub-cellular impact on the larval gut was also evaluated. First/third instar larvae or the adult Drosophila were exposed orally to green fluorescence emitting CdTe QDs (0.2-100 μM), and organisms' longevity, emergence, reproductive performance, locomotion, and reactive oxygen species (ROS), and cell death were assessed. Uptake of semiconductor CdTe QDs was observed as green fluorescence in the gut. A significant decline in percentage survivability up to 80% was evident at high CdTe QDs concentrations (25 and 100 μM). The developmental toxicity was marked by delayed and reduced fly emergence after CdTe exposure. The teratogenic effect was evident with significant wing deformities at 25 and 100 μM concentrations. However, at the reproductive level, adult flies' fecundity, fertility, and hatchability were highly affected even at low concentrations (1 μM). Surprisingly, the climbing ability of Drosophila was unaffected at any of the used CdTe QDs concentrations. In addition to organismal toxicity, the ROS level and cell death were elevated in gut cells, confirming the sub-cellular toxicity of CdTe QDs. Furthermore, we observed a significant rescue in CdTe QDs-associated developmental, reproductive, and survival adversities when organisms were co-exposed with N-acetyl-cysteine (NAC, an antioxidant) and CdTe QDs. Overall, our findings indicate that the environmental release of aqueously dispersible CdTe QDs raises a long-lasting health concern on the development, reproduction, and survivability of an organism.
Collapse
Affiliation(s)
- Jagdish Gopal Paithankar
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Sharanya Kushalan
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Nijil S
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Nanobiotechnology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Smitha Hegde
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India
| | - Sudarshan Kini
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Nanobiotechnology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| | - Anurag Sharma
- Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Kotekar-Beeri Road, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
14
|
Chang B, Li D, Ren Y, Qu C, Shi X, Liu R, Liu H, Tian J, Hu Z, Sun T, Cheng Z. A phosphorescent probe for in vivo imaging in the second near-infrared window. Nat Biomed Eng 2022; 6:629-639. [PMID: 34385694 DOI: 10.1038/s41551-021-00773-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
In the second near-infrared spectral window (NIR-II; with wavelengths of 1,000-1,700 nm), in vivo fluorescence imaging can take advantage of reduced tissue autofluorescence and lower light absorption and scattering by tissue. Here, we report the development and in vivo application of a NIR-II phosphorescent probe that has lifetimes of hundreds of microseconds and a Stokes shift of 430 nm. The probe is made of glutathione-capped copper-indium-selenium nanotubes, and in acidic environments (pH 5.5-6.5) switches from displaying fluorescence to phosphorescence. In xenograft models of osteosarcoma and breast cancer, intravenous or intratumoral injections of the probe enabled phosphorescence imaging at signal-to-background ratios, spatial resolutions and sensitivities higher than NIR-II fluorescence imaging with polymer-stabilized copper-indium-sulfide nanorods. Phosphorescence imaging may offer superior imaging performance for a range of biomedical uses.
Collapse
Affiliation(s)
- Baisong Chang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, People's Republic of China.,Molecular Imaging Program at Stanford (MIPS), Bio-X Program and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Daifeng Li
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA.,Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Ying Ren
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA
| | - Chunrong Qu
- Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Xiaojing Shi
- Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ruiqi Liu
- Institute of Molecular Medicine, College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Hongguang Liu
- Institute of Molecular Medicine, College of Life and Health Sciences, Northeastern University, Shenyang, People's Republic of China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Zhenhua Hu
- Key Laboratory of Molecular Imaging, State Key Laboratory of Management and Control for Complex Systems, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, People's Republic of China.
| | - Taolei Sun
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, People's Republic of China.
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Bio-X Program and Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University, Stanford, CA, USA. .,Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
15
|
Wang M, Hu C, Su Q. Luminescent Lifetime Regulation of Lanthanide-Doped Nanoparticles for Biosensing. BIOSENSORS 2022; 12:131. [PMID: 35200391 PMCID: PMC8869906 DOI: 10.3390/bios12020131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/09/2022] [Accepted: 02/12/2022] [Indexed: 05/16/2023]
Abstract
Lanthanide-doped nanoparticles possess numerous advantages including tunable luminescence emission, narrow peak width and excellent optical and thermal stability, especially concerning the long lifetime from microseconds to milliseconds. Differing from other shorter-lifetime fluorescent nanomaterials, the long lifetime of lanthanide-doped nanomaterials is independent with background fluorescence interference and biological tissue depth. This review presents the recent advances in approaches to regulating the lifetime and applications of bioimaging and biodetection. We begin with the introduction of the strategies for regulating the lifetime by modulating the core-shell structure, adjusting the concentration of sensitizer and emitter, changing energy transfer channel, establishing a fluorescence resonance energy transfer pathway and changing temperature. We then summarize the applications of these nanoparticles in biosensing, including ion and molecule detecting, DNA and protease detection, cell labeling, organ imaging and thermal and pH sensing. Finally, the prospects and challenges of the lanthanide lifetime regulation for fundamental research and practical applications are also discussed.
Collapse
Affiliation(s)
- Mingkai Wang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China;
| | - Qianqian Su
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| |
Collapse
|
16
|
Xue D, Zou W, Liu D, Li L, Chen T, Yang Z, Chen Y, Wang X, Lu W, Lin G. Cytotoxicity and transcriptome changes triggered by CuInS 2/ZnS quantum dots in human glial cells. Neurotoxicology 2021; 88:134-143. [PMID: 34785253 DOI: 10.1016/j.neuro.2021.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 11/03/2021] [Accepted: 11/12/2021] [Indexed: 01/27/2023]
Abstract
As a newly developed cadmium-free quantum dot (QD), CuInS2/ZnS has great application potential in many fields, but its biological safety has not been fully understood. In this study, the in vitro toxicity of CuInS2/ZnS QDs on U87 human glioma cell line was explored. The cells were treated with different concentrations of QDs (12.5, 25, 50 and 100 μg/mL), and the uptake of QDs by the U87 cells was detected by fluorescence imaging and flow cytometry. The cell viability was observed by MTT assay, and the gene expression profile was analyzed by transcriptome sequencing. These results showed that QDs could enter the cells and mainly located in the cytoplasm. The uptake rate was over 90 % when the concentration of QDs reached 25 μg/mL. The cell viability (50 and 100 μg/mL) increased at 24 h (P < 0.05), but no significant difference after 48 h and 72 h treatment. The results of differential transcription showed that coding RNA accounted for the largest proportion (62.15 %), followed by long non-coding RNA (18.65 %). Total 220 genes were up-regulated and 1515 genes were down-regulated, and significantly altered gene functions included nucleosome, chromosome-DNA binding, and chromosome assembly. In conclusion, CuInS2/ZnS QDs could enter U87 cells, did not reduce the cell viability, but would obviously alter the gene expression profile. These findings provide valuable information for a proper understanding of the toxicity risk of CuInS2/ZnS QD and promote the rational utilization of QDs in the future.
Collapse
Affiliation(s)
- Dahui Xue
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen, China; School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Wenyi Zou
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Dongmeng Liu
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen, China; School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Li Li
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen, China; School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Tingting Chen
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhiwen Yang
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yajing Chen
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Xiaomei Wang
- School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Wencan Lu
- Department of Spine Surgery, Shenzhen University General Hospital, Shenzhen, 518060, China.
| | - Guimiao Lin
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen, China; School of Basic Medical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
17
|
Liang Z, Khawar MB, Liang J, Sun H. Bio-Conjugated Quantum Dots for Cancer Research: Detection and Imaging. Front Oncol 2021; 11:749970. [PMID: 34745974 PMCID: PMC8569511 DOI: 10.3389/fonc.2021.749970] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/30/2021] [Indexed: 12/20/2022] Open
Abstract
Ultrasound, computed tomography, magnetic resonance, and gamma scintigraphy-based detection and bio-imaging technologies have achieved outstanding breakthroughs in recent years. However, these technologies still encounter several limitations such as insufficient sensitivity, specificity and security that limit their applications in cancer detection and bio-imaging. The semiconductor quantum dots (QDs) are a kind of newly developed fluorescent nanoparticles that have superior fluorescence intensity, strong resistance to photo-bleaching, size-tunable light emission and could produce multiple fluorescent colors under single-source excitation. Furthermore, QDs have optimal surface to link with multiple targets such as antibodies, peptides, and several other small molecules. Thus, QDs might serve as potential, more sensitive and specific methods of detection than conventional methods applied in cancer molecular targeting and bio-imaging. However, many challenges such as cytotoxicity and nonspecific uptake still exist limiting their wider applications. In the present review, we aim to summarize the current applications and challenges of QDs in cancer research mainly focusing on tumor detection, bio-imaging, and provides opinions on how to address these challenges.
Collapse
Affiliation(s)
- Zhengyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China
| | - Muhammad Babar Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China.,Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences, University of Central Punjab, Lahore, Pakistan
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China
| |
Collapse
|
18
|
Abbasi Kajani A, Haghjooy Javanmard S, Asadnia M, Razmjou A. Recent Advances in Nanomaterials Development for Nanomedicine and Cancer. ACS APPLIED BIO MATERIALS 2021; 4:5908-5925. [PMID: 35006909 DOI: 10.1021/acsabm.1c00591] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cancer is considered one of the leading causes of death, with a growing number of cases worldwide. However, the early diagnosis and efficient therapy of cancer have remained a critical challenge. The emergence of nanomedicine has opened up a promising window to address the drawbacks of cancer detection and treatment. A wide range of engineered nanomaterials and nanoplatforms with different shapes, sizes, and composition has been developed for various biomedical applications. Nanomaterials have been increasingly used in various applications in bioimaging, diagnosis, and therapy of cancers. Recently, numerous multifunctional and smart nanoparticles with the ability of simultaneous diagnosis and targeted cancer therapy have been reported. The multidisciplinary attempts led to the development of several exciting clinically approved nanotherapeutics. The nanobased materials and devices have also been used extensively to develop point-of-care and highly sensitive methods of cancer detection. In this review article, the most significant achievements and latest advances in the nanomaterials development for cancer nanomedicine are critically discussed. In addition, the future perspectives of this field are evaluated.
Collapse
Affiliation(s)
- Abolghasem Abbasi Kajani
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746-73461, Iran
| | - Mohsen Asadnia
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Amir Razmjou
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan 73441-81746, Iran
- UNESCO Centre for Membrane Science and Technology, School of Chemical Engineering, University of New South Wales, Sydney, New South Wales 2052, Australia
- Centre for Technology in Water and Wastewater, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
19
|
Broadwater D, Medeiros HCD, Lunt RR, Lunt SY. Current Advances in Photoactive Agents for Cancer Imaging and Therapy. Annu Rev Biomed Eng 2021; 23:29-60. [PMID: 34255992 DOI: 10.1146/annurev-bioeng-122019-115833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Photoactive agents are promising complements for both early diagnosis and targeted treatment of cancer. The dual combination of diagnostics and therapeutics is known as theranostics. Photoactive theranostic agents are activated by a specific wavelength of light and emit another wavelength, which can be detected for imaging tumors, used to generate reactive oxygen species for ablating tumors, or both. Photodynamic therapy (PDT) combines photosensitizer (PS) accumulation and site-directed light irradiation for simultaneous imaging diagnostics and spatially targeted therapy. Although utilized since the early 1900s, advances in the fields of cancer biology, materials science, and nanomedicine have expanded photoactive agents to modern medical treatments. In this review we summarize the origins of PDT and the subsequent generations of PSs and analyze seminal research contributions that have provided insight into rational PS design, such as photophysics, modes of cell death, tumor-targeting mechanisms, and light dosing regimens. We highlight optimizable parameters that, with further exploration, can expand clinical applications of photoactive agents to revolutionize cancer diagnostics and treatment.
Collapse
Affiliation(s)
- Deanna Broadwater
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Hyllana C D Medeiros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA
| | - Richard R Lunt
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824, USA; , .,Department of Physics and Astronomy, Michigan State University, East Lansing, Michigan 48824, USA
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, USA.,Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48824, USA; ,
| |
Collapse
|
20
|
Desale K, Kuche K, Jain S. Cell-penetrating peptides (CPPs): an overview of applications for improving the potential of nanotherapeutics. Biomater Sci 2021; 9:1153-1188. [PMID: 33355322 DOI: 10.1039/d0bm01755h] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the field of nanotherapeutics, gaining cellular entry into the cytoplasm of the target cell continues to be an ultimate challenge. There are many physicochemical factors such as charge, size and molecular weight of the molecules and delivery vehicles, which restrict their cellular entry. Hence, to dodge such situations, a class of short peptides called cell-penetrating peptides (CPPs) was brought into use. CPPs can effectively interact with the cell membrane and can assist in achieving the desired intracellular entry. Such strategy is majorly employed in the field of cancer therapy and diagnosis, but now it is also used for other purposes such as evaluation of atherosclerotic plaques, determination of thrombin levels and HIV therapy. Thus, the current review expounds on each of these mentioned aspects. Further, the review briefly summarizes the basic know-how of CPPs, their utility as therapeutic molecules, their use in cancer therapy, tumor imaging and their assistance to nanocarriers in improving their membrane penetrability. The review also discusses the challenges faced with CPPs pertaining to their stability and also mentions the strategies to overcome them. Thus, in a nutshell, this review will assist in understanding how CPPs can present novel possibilities for resolving the conventional issues faced with the present-day nanotherapeutics.
Collapse
Affiliation(s)
- Kalyani Desale
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| | - Kaushik Kuche
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab-160062, India.
| |
Collapse
|
21
|
Hurtado de Mendoza T, Mose ES, Botta GP, Braun GB, Kotamraju VR, French RP, Suzuki K, Miyamura N, Teesalu T, Ruoslahti E, Lowy AM, Sugahara KN. Tumor-penetrating therapy for β5 integrin-rich pancreas cancer. Nat Commun 2021; 12:1541. [PMID: 33750829 PMCID: PMC7943581 DOI: 10.1038/s41467-021-21858-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by marked desmoplasia and drug resistance due, in part, to poor drug delivery to extravascular tumor tissue. Here, we report that carcinoma-associated fibroblasts (CAFs) induce β5 integrin expression in tumor cells in a TGF-β dependent manner, making them an efficient drug delivery target for the tumor-penetrating peptide iRGD. The capacity of iRGD to deliver conjugated and co-injected payloads is markedly suppressed when β5 integrins are knocked out in the tumor cells. Of note, β5 integrin knock-out in tumor cells leads to reduced disease burden and prolonged survival of the mice, demonstrating its contribution to PDAC progression. iRGD significantly potentiates co-injected chemotherapy in KPC mice with high β5 integrin expression and may be a powerful strategy to target an aggressive PDAC subpopulation.
Collapse
Affiliation(s)
| | - Evangeline S Mose
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Gregory P Botta
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, Division of Hematology/Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Department of Molecular Medicine, Scripps Research Translational Institute, La Jolla, CA, USA
| | - Gary B Braun
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Venkata R Kotamraju
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Randall P French
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Kodai Suzuki
- Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Norio Miyamura
- Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Tambet Teesalu
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
- Laboratory of Cancer Biology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| | - Kazuki N Sugahara
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
- Department of Surgery, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
22
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
23
|
Tan Y, He K, Tang B, Chen H, Zhao Z, Zhang C, Lin L, Liu J. Precisely Regulated Luminescent Gold Nanoparticles for Identification of Cancer Metastases. ACS NANO 2020; 14:13975-13985. [PMID: 32865989 DOI: 10.1021/acsnano.0c06388] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The nanoprobes for identification of cancer metastases in the mononuclear phagocyte system (MPS) organs are of significant importance but are limited due to the long-standing challenge of low tumor-targeting specificity with inadequate targeting efficiency and high nonspecific accumulation. Here, we report a surface regulation strategy that integrates the tumor-acidity-activated charge-reversal behavior and precise control in both hydrodynamic diameter (HD) and surface charge on ultrasmall luminescent gold nanoparticles (AuNPs) to achieve significantly high tumor-targeting specificity. The precise regulation of AuNPs to a rational HD and surface charge could rapidly and selectively recognize small metastatic tumors (∼1 mm) in liver and lung with high signal-to-noise ratios of 4.6 and 4.5, respectively. These results help further understand the in vivo transport of nanoprobes and provide guidance for design of translatable nanosized nanomedicines in cancer metastasis theranostics.
Collapse
Affiliation(s)
- Yue Tan
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Kui He
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Bing Tang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Huarui Chen
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Zhipeng Zhao
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Chengqian Zhang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Li Lin
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jinbin Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
24
|
Tobi A, Willmore AA, Kilk K, Sidorenko V, Braun GB, Soomets U, Sugahara KN, Ruoslahti E, Teesalu T. Silver Nanocarriers Targeted with a CendR Peptide Potentiate the Cytotoxic Activity of an Anticancer Drug. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Allan Tobi
- Laboratory of Cancer Biology Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
| | - Anne‐Mari A. Willmore
- Laboratory of Cancer Biology Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
| | - Kalle Kilk
- Department of Biochemistry Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
| | - Valeria Sidorenko
- Laboratory of Cancer Biology Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
| | - Gary B. Braun
- Cancer Research Center Sanford‐Burnham‐Prebys Medical Discovery Institute 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Ursel Soomets
- Department of Biochemistry Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
| | - Kazuki N. Sugahara
- Department of Surgery Columbia University College of Physicians and Surgeons New York NY 10032 USA
| | - Erkki Ruoslahti
- Cancer Research Center Sanford‐Burnham‐Prebys Medical Discovery Institute 10901 North Torrey Pines Road La Jolla CA 92037 USA
| | - Tambet Teesalu
- Laboratory of Cancer Biology Institute of Biomedicine and Translational Medicine University of Tartu Ravila 14b Tartu 50411 Estonia
- Cancer Research Center Sanford‐Burnham‐Prebys Medical Discovery Institute 10901 North Torrey Pines Road La Jolla CA 92037 USA
- Center for Nanomedicine and Department of Cell Molecular and Developmental Biology University of California Santa Barbara Santa Barbara La Jolla CA 93106 USA
| |
Collapse
|
25
|
Halloran D, Vrathasha V, Durbano HW, Nohe A. Bone Morphogenetic Protein-2 Conjugated to Quantum Dot ®s is Biologically Functional. NANOMATERIALS 2020; 10:nano10061208. [PMID: 32575709 PMCID: PMC7353091 DOI: 10.3390/nano10061208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/12/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
Quantum Dot®s (QDot®s) are novel, semi-conductive nanostructures that emit a certain fluorescence when excited by specific wavelengths. QDot®s are more photostable, brighter, and photobleach less than other fluorescent dyes. These characteristics give them the potential to be used in many biological applications. The shells of QDot®s are coated with functional groups, such as carboxylate and organic groups, allowing them to couple to peptides/proteins and be used for real-time imaging and high-resolution microscopy. Here, we utilize Quantum Dot®s and Bone Morphogenetic Protein-2 (BMP-2) to create a BMP-2-QDot®s conjugate. BMP-2 is a growth factor that drives many processes such as cardiogenesis, neural growth, and osteogenesis. Despite its numerous roles, the trafficking and uptake of BMP-2 into cells is not well-established, especially during progression of diseases. The results presented here demonstrate for the first time a fluorescent BMP-2 analog that binds to the BMP-receptors (BMPRs), remains biologically active, and is stable for long time periods. Previous attempts to develop a biological BMP-2 analog with Fluorescein isothiocyanate (FITC) or nanodiamonds lacked data on the analog’s stability. Furthermore, these analogs did not address whether they can signal within the cell by binding to the BMPRs or were mediated by non-stable conjugates.
Collapse
Affiliation(s)
| | | | | | - Anja Nohe
- Correspondence: ; Tel.: +1-302-831-6977
| |
Collapse
|
26
|
Tang T, Wei Y, Yang Q, Yang Y, Sailor MJ, Pang HB. Rapid chelator-free radiolabeling of quantum dots for in vivo imaging. NANOSCALE 2019; 11:22248-22254. [PMID: 31746913 DOI: 10.1039/c9nr08508d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Most current nanoparticle-based PET tracers are radiolabeled through metal chelators conjugated on the nanoparticle surface. Metal chelation usually requires sophisticated optimization and may impact the physical or chemical properties of nanoparticles, which leads to the changes in their distribution and pharmacokinetics in vivo. A chelator-free radiolabeling approach is thus highly desirable. Here, we report that zinc sulfide (ZnS) quantum dots (QDs) can be rapidly radiolabeled with 68Ga or 64Cu through cation exchange without chelators. The radiolabeling was accomplished in times as short as 5 min at 37 °C in aqueous solution, yielding a high labeling efficiency and radiochemical purity for both isotopes. Surface functionalization with targeting peptides was also readily achieved to enable or enhance the cellular uptake of QDs. In vivo PET imaging showed that 64Cu-labeled QDs had a much higher tumor uptake (7.3% ID g-1) than 64Cu-DOTA in a murine cancer model. Overall, this study presents a QD-based platform to achieve convenient and chelator-free radiolabeling, and improve PET imaging of solid tumors.
Collapse
Affiliation(s)
- Tang Tang
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Wu L, Wang Y, He R, Zhang Y, He Y, Wang C, Lu Z, Liu Y, Ju H. Fluorescence hydrogel array based on interfacial cation exchange amplification for highly sensitive microRNA detection. Anal Chim Acta 2019; 1080:206-214. [DOI: 10.1016/j.aca.2019.07.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/09/2019] [Accepted: 07/11/2019] [Indexed: 10/26/2022]
|
28
|
Yang L, Sun Z, Zhang L, Cai Y, Peng Y, Cao T, Zhang Y, Lu H. Chemical labeling for fine mapping of IgG N-glycosylation by ETD-MS. Chem Sci 2019; 10:9302-9307. [PMID: 32110292 PMCID: PMC7006626 DOI: 10.1039/c9sc02491c] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/21/2019] [Indexed: 12/25/2022] Open
Abstract
Immunoglobulin G (IgG), which contains four subclasses (IgG1-4), is one of the most important classes of glycoproteins in the immune system. Because of its importance in the immune system, a steady increase of interest in developing IgG as the biomarker or biotherapeutic agent for the treatment of diseases has been seen, as most therapeutic mAbs were IgG-based. N-Glycosylation of IgG is crucial for its effector function and makes IgG highly heterogeneous both in structure and function, although all four subclasses of IgG contain only a single N-glycosylation site in the Fc region with a highly similar amino acid sequence. Therefore, fine mapping of IgG glycosylation is necessary for understanding the IgG function and avoiding aberrant glycosylation in mAbs. However, site-specific and comprehensive N-glycosylation analysis of IgG subclasses still cannot be achieved by MS alone due to the partial sequence coverage and loss of connections among glycosylation of the protein sequence. We report here a chemical labeling strategy to improve the electron transfer dissociation efficiency in mass spectrometry analysis, which enables a 100% peptide sequence coverage of N-glycopeptides in all subclasses of IgG. Combined with high-energy collisional dissociation for the fragmentation of glycans, fine mapping of the N-glycosylation profile of IgG is achieved. This comprehensive glycosylation analysis strategy for the first time allows the discrimination of IgG3 and IgG4 intact N-glycopeptides with high similarity in sequence without the antibody-based pre-separation. Using this strategy, aberrant serum IgG N-glycosylation for four IgG subclasses associated with cirrhosis and hepatocellular carcinoma was revealed. Moreover, this method identifies 5 times more intact glycopeptides from human serum than the native-ETD method, implying that the approach can also accommodate large-scale site-specific profiling of glycoproteomes.
Collapse
Affiliation(s)
- Lijun Yang
- Shanghai Cancer Center , Department of Chemistry , Fudan University , Shanghai 200032 , China . ;
| | - Zhenyu Sun
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| | - Lei Zhang
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| | - Yan Cai
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| | - Ye Peng
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| | - Ting Cao
- Shanghai Cancer Center , Department of Chemistry , Fudan University , Shanghai 200032 , China . ;
| | - Ying Zhang
- Shanghai Cancer Center , Department of Chemistry , Fudan University , Shanghai 200032 , China . ;
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| | - Haojie Lu
- Shanghai Cancer Center , Department of Chemistry , Fudan University , Shanghai 200032 , China . ;
- Institutes of Biomedical Sciences , NHC Key Laboratory of Glycoconjugates Research , Fudan University , Shanghai 200032 , China
| |
Collapse
|
29
|
Uchida S. Frontiers and progress in cation-uptake and exchange chemistry of polyoxometalate-based compounds. Chem Sci 2019; 10:7670-7679. [PMID: 31803405 PMCID: PMC6839602 DOI: 10.1039/c9sc02823d] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cation-uptake and exchange has been an important topic in both basic and applied chemistry relevant to life and materials science. For example, living cells contain appreciable amounts of Na+ and K+, and their concentrations are regulated by the sodium-potassium pump. Solid-state cation-exchangers such as clays and zeolites both natural and synthetic have been used widely in water softening and purification, separation of metal ions and biomolecules, etc. Polyoxometalates (POMs) are robust, discrete, and structurally well-defined metal-oxide cluster anions, and have stimulated research in broad fields of sciences. In this perspective, cation-uptake and exchange in POM and POM-based compounds are categorized and reviewed in three groups: (i) POMs as inorganic crown ethers and cryptands, (ii) POM-based ionic solids as cation-exchangers, and (iii) reduction-induced cation-uptake in POM-based ionic solids, which is based on a feature of POMs that they are redox-active and multi-electron transfer occurs reversibly in multiple steps. This method can be utilized to synthesize mixed-valence metal clusters in metal ion-exchanged POM-based ionic solids.
Collapse
Affiliation(s)
- Sayaka Uchida
- Department of Basic Science , School of Arts and Sciences , The University of Tokyo , Komaba 3-8-1, Meguro-ku , Tokyo 153-8902 , Japan .
| |
Collapse
|
30
|
Wagner AM, Knipe JM, Orive G, Peppas NA. Quantum dots in biomedical applications. Acta Biomater 2019; 94:44-63. [PMID: 31082570 PMCID: PMC6642839 DOI: 10.1016/j.actbio.2019.05.022] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/10/2019] [Accepted: 05/08/2019] [Indexed: 01/14/2023]
Abstract
Semiconducting nanoparticles, more commonly known as quantum dots, possess unique size and shape dependent optoelectronic properties. In recent years, these unique properties have attracted much attention in the biomedical field to enable real-time tissue imaging (bioimaging), diagnostics, single molecule probes, and drug delivery, among many other areas. The optical properties of quantum dots can be tuned by size and composition, and their high brightness, resistance to photobleaching, multiplexing capacity, and high surface-to-volume ratio make them excellent candidates for intracellular tracking, diagnostics, in vivo imaging, and therapeutic delivery. We discuss recent advances and challenges in the molecular design of quantum dots are discussed, along with applications of quantum dots as drug delivery vehicles, theranostic agents, single molecule probes, and real-time in vivo deep tissue imaging agents. We present a detailed discussion of the biodistribution and toxicity of quantum dots, and highlight recent advances to improve long-term stability in biological buffers, increase quantum yield following bioconjugation, and improve clearance from the body. Last, we present an outlook on future challenges and strategies to further advance translation to clinical application. STATEMENT OF SIGNIFICANCE: Semiconducting nanoparticles, commonly known as quantum dots, possess unique size and shape dependent electrical and optical properties. In recent years, they have attracted much attention in biomedical imaging to enable diagnostics, single molecule probes, and real-time imaging of tumors. This review discusses recent advances and challenges in the design of quantum dots, and highlights how these strategies can further advance translation to clinical applications.
Collapse
Affiliation(s)
- Angela M Wagner
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Jennifer M Knipe
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Nicholas A Peppas
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA; Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA; Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
31
|
Yahyaei B, Pourali P. One step conjugation of some chemotherapeutic drugs to the biologically produced gold nanoparticles and assessment of their anticancer effects. Sci Rep 2019; 9:10242. [PMID: 31308430 PMCID: PMC6629879 DOI: 10.1038/s41598-019-46602-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 07/02/2019] [Indexed: 11/23/2022] Open
Abstract
Recent research tried to analyze the conjugation of some chemotherapeutic drugs to the biologically produced gold nanoparticles (GNPs) in one step, without the use of any additional linkers. GNPs was produced using Fusarium oxysporum and their presence was confirmed using spectrophotometer, transmission electron microscope (TEM), X-ray diffraction (XRD) and fourier transform infrared (FTIR) analyses. In order to carry out the conjugation study, capecitabine, tamoxifen, and paclitaxel were added dropwise to the GNPs solution under stirring condition and spectrophotometer, dynamic light scattering (DLS) and FTIR analyses were performed to prove the successful conjugation. Finally, AGS and MCF7 cell lines were used for methyl thiazol tetrazolium (MTT) assay to determine the toxicity of each drug and its conjugated form. Results showed that the spherical and hexagonal GNPs with maximum absorbance peak around 524 nm and average sizes less than 20 nm were produced. FTIR analysis clarified the presence of proteins on the surfaces of the GNPs. After the conjugation process although the FTIR analysis demonstrated that all the drugs were successfully conjugated to GNPs, MTT assay revealed that unlike the paclitaxel conjugated GNPs, capecitabine and tamoxifen conjugates displayed no toxic effects due to their deactivation and low half-lives. Moreover the average size and polydispersity index (PDI) of the GNPs after conjugation with all the three tested drugs increased. In conclusion different types of drugs could conjugate to the GNPs but it is important to employ high stable forms of the drugs in the conjugation procedure.
Collapse
Affiliation(s)
- Behrooz Yahyaei
- Department of Medical Sciences, Shahrood Branch, Islamic Azad University, Shahrood, Iran.,Biological Nanoparticles in Medicine Research Center, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Parastoo Pourali
- Department of Medical Sciences, Shahrood Branch, Islamic Azad University, Shahrood, Iran. .,Biological Nanoparticles in Medicine Research Center, Shahrood Branch, Islamic Azad University, Shahrood, Iran.
| |
Collapse
|
32
|
Zhang LJ, Xia L, Xie HY, Zhang ZL, Pang DW. Quantum Dot Based Biotracking and Biodetection. Anal Chem 2018; 91:532-547. [DOI: 10.1021/acs.analchem.8b04721] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Li-Juan Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Li Xia
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P.R. China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, State Key Laboratory of Virology, The Institute for Advanced Studies, and Wuhan Institute of Biotechnology, Wuhan University, Luojia Hill, Wuhan 430072, P.R. China
- College of Chemistry, Nankai University, 94 Weijin Road, Tianjin 300071, P.R. China
| |
Collapse
|
33
|
Near-infrared MnCuInS/ZnS@BSA and urchin-like Au nanoparticle as a novel donor-acceptor pair for enhanced FRET biosensing. Anal Chim Acta 2018; 1042:71-78. [DOI: 10.1016/j.aca.2018.05.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 04/20/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022]
|
34
|
Zhang L, Lin Z, Yu YX, Jiang BP, Shen XC. Multifunctional hyaluronic acid-derived carbon dots for self-targeted imaging-guided photodynamic therapy. J Mater Chem B 2018; 6:6534-6543. [PMID: 32254861 DOI: 10.1039/c8tb01957f] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
It is of vital importance to engineer the surface structures of carbon dots (CDs) to satisfy their practical biomedical applications, including imaging and treatment. In this work, one type of hyaluronic acid-derived CD (HA-CD) was synthesized via a facile one-step hydrothermal method using cancer cell-targeted HA as a precursor. The as-prepared HA-CDs were targeted actively toward CD44 receptor-overexpressing cancer cells because a partial HA structure remained on the HA-CD surface. Beyond this, HA-CDs can act as a novel photosensitizer, because they can generate O2˙- under 650 nm laser irradiation, and they also exhibit excellent blue photoluminescence emission. The in vitro results revealed that HA-CDs imaged selectively CD44-overexpressing cancer cells and inhibited their growth under 650 nm laser irradiation. Thus, HA-CDs can serve as a promising self-targeted imaging-guided photodynamic therapy (PDT) agent for cancer. The present research provides a promising new method to simply construct multifunctional CD-based targeted phototheranostic systems.
Collapse
Affiliation(s)
- Lizhen Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, P. R. China.
| | | | | | | | | |
Collapse
|
35
|
Yuan Z, Gui L, Zheng J, Chen Y, Qu S, Shen Y, Wang F, Er M, Gu Y, Chen H. GSH-Activated Light-Up Near-Infrared Fluorescent Probe with High Affinity to α vβ 3 Integrin for Precise Early Tumor Identification. ACS APPLIED MATERIALS & INTERFACES 2018; 10:30994-31007. [PMID: 30141897 DOI: 10.1021/acsami.8b09841] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The development of tumor-associated, stimuli-driven, turn-on near-infrared (NIR) fluorophores requires urgent attention because of their potential in selective and precise tumor diagnosis. Herein, we describe a NIR fluorescent probe (CyA-cRGD) comprised of a fluorescence reporting unit (a cyanine dye) linked with a GSH-responsive unit (nitroazo aryl ether group) and a tumor-targeting unit (cRGD). The NIR fluorescence of CyA-cRGD with sensitive and selective response to GSH can act as a direct off-on signal reporter for GSH monitoring. Notably, CyA-cRGD possesses improved biocompatibility compared with CyA, which is highly desirable for in vivo fluorescence tracking of cancer. Confocal fluorescence imaging confirmed the tumor-targeting capability and GSH detection ability of CyA-cRGD in tumor cells, normal cells, and coincubated tumor /normal cells and in the three-dimensional multicellular tumor spheroid. Furthermore, it was validated that CyA-cRGD could detect tumor precisely in GSH and integrin αvβ 3 high-expressed tumor-bearing mouse models. Importantly, it was confirmed that CyA-cRGD possessed high efficiency for early-stage tumor imaging in mouse models with tumor cells implanted within 72 h. This method provided significant advances toward more in-depth understanding and exploration of tumor imaging, which may potentially be applied for clinical early tumor diagnosis.
Collapse
Affiliation(s)
- Zhenwei Yuan
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Lijuan Gui
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Jinrong Zheng
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Yisha Chen
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Sisi Qu
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Yuanzhi Shen
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Fei Wang
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Murat Er
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Yueqing Gu
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| | - Haiyan Chen
- Department of Biomedical Engineering, School of Engineering , China Pharmaceutical University , 24 Tongjia Lane , Gulou District, Nanjing 210009 , China
| |
Collapse
|
36
|
Mangeolle T, Yakavets I, Marchal S, Debayle M, Pons T, Bezdetnaya L, Marchal F. Fluorescent Nanoparticles for the Guided Surgery of Ovarian Peritoneal Carcinomatosis. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E572. [PMID: 30050022 PMCID: PMC6116267 DOI: 10.3390/nano8080572] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 07/20/2018] [Accepted: 07/22/2018] [Indexed: 01/07/2023]
Abstract
Complete surgical resection is the ideal cure for ovarian peritoneal carcinomatosis, but remains challenging. Fluorescent guided surgery can be a promising approach for precise cytoreduction when appropriate fluorophore is used. In the presence paper, we review already developed near- and short-wave infrared fluorescent nanoparticles, which are currently under investigation for peritoneal carcinomatosis fluorescence imaging. We also highlight the main ways to improve the safety of nanoparticles, for fulfilling prerequisites of clinical application.
Collapse
Affiliation(s)
- Tristan Mangeolle
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France.
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| | - Ilya Yakavets
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France.
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
- Laboratory of Biophysics and Biotechnology, Belarusian State University, 4 Nezavisimosti Avenue, 220030 Minsk, Belarus.
| | - Sophie Marchal
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France.
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| | - Manon Debayle
- LPEM, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université, 75005 Paris, France.
| | - Thomas Pons
- LPEM, ESPCI Paris, PSL Research University, CNRS, Sorbonne Université, 75005 Paris, France.
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France.
- Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| | - Frédéric Marchal
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France.
- Surgical Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
37
|
Wang G, Li Z, Ma N. Next-Generation DNA-Functionalized Quantum Dots as Biological Sensors. ACS Chem Biol 2018; 13:1705-1713. [PMID: 29257662 DOI: 10.1021/acschembio.7b00887] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
DNA-functionalized quantum dots (DNA-QDs) have found considerable application in biosensing and bioimaging. Different from the first generation (I-G) DNA-QDs prepared via conventional bioconjugation chemistry, the second generation (II-G) DNA-QDs prepared via one-step DNA-templated QD synthesis features a defined number of DNA valencies (usually monovalency), which is preferable for controlled assembly and biological targeting. In this review, we summarize recent progress in designing QD probes based on II-G DNA-QDs for advanced sensing and imaging applications. It opens up new avenues for highly sensitive and intelligent sensing of a range of disease-relevant biomolecules in vitro and in living cells.
Collapse
Affiliation(s)
- Ganglin Wang
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, People’s Republic of China
| | - Zhi Li
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, People’s Republic of China
| | - Nan Ma
- The Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, People’s Republic of China
| |
Collapse
|
38
|
Simón-Gracia L, Hunt H, Teesalu T. Peritoneal Carcinomatosis Targeting with Tumor Homing Peptides. Molecules 2018; 23:molecules23051190. [PMID: 29772690 PMCID: PMC6100015 DOI: 10.3390/molecules23051190] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/08/2018] [Accepted: 05/10/2018] [Indexed: 12/16/2022] Open
Abstract
Over recent decades multiple therapeutic approaches have been explored for improved management of peritoneally disseminated malignancies—a grim condition known as peritoneal carcinomatosis (PC). Intraperitoneal (IP) administration can be used to achieve elevated local concentration and extended half-life of the drugs in the peritoneal cavity to improve their anticancer efficacy. However, IP-administered chemotherapeutics have a short residence time in the IP space, and are not tumor selective. An increasing body of work suggests that functionalization of drugs and nanoparticles with targeting peptides increases their peritoneal retention and provides a robust and specific tumor binding and penetration that translates into improved therapeutic response. Here we review the progress in affinity targeting of intraperitoneal anticancer compounds, imaging agents and nanoparticles with tumor-homing peptides. We review classes of tumor-homing peptides relevant for PC targeting, payloads for peptide-guided precision delivery, applications for targeted compounds, and the effects of nanoformulation of drugs and imaging agents on affinity-based tumor delivery.
Collapse
Affiliation(s)
- Lorena Simón-Gracia
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia.
| | - Hedi Hunt
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia.
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia.
- Cancer Research Center, Sanford-Burnham-Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
39
|
Villa I, Villa C, Monguzzi A, Babin V, Tervoort E, Nikl M, Niederberger M, Torrente Y, Vedda A, Lauria A. Demonstration of cellular imaging by using luminescent and anti-cytotoxic europium-doped hafnia nanocrystals. NANOSCALE 2018; 10:7933-7940. [PMID: 29671445 DOI: 10.1039/c8nr00724a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Luminescent nanoparticles are researched for their potential impact in medical science, but no materials approved for parenteral use have been available so far. To overcome this issue, we demonstrate that Eu3+-doped hafnium dioxide nanocrystals can be used as non-toxic, highly stable probes for cellular optical imaging and as radiosensitive materials for clinical treatment. Furthermore, viability and biocompatibility tests on artificially stressed cell cultures reveal their ability to buffer reactive oxygen species, proposing an anti-cytotoxic feature interesting for biomedical applications.
Collapse
Affiliation(s)
- Irene Villa
- Dipartimento di Scienza dei Materiali, Università degli Studi Milano Bicocca, via R. Cozzi 55, 20125 Milano, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
A cancer vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat Commun 2018; 9:1532. [PMID: 29670088 PMCID: PMC5906566 DOI: 10.1038/s41467-018-03915-4] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 03/22/2018] [Indexed: 12/27/2022] Open
Abstract
Vaccines to induce effective and sustained antitumor immunity have great potential for postoperative cancer therapy. However, a robust cancer vaccine simultaneously eliciting tumor-specific immunity and abolishing immune resistance continues to be a challenge. Here we present a personalized cancer vaccine (PVAX) for postsurgical immunotherapy. PVAX is developed by encapsulating JQ1 (a BRD4 inhibitor) and indocyanine green (ICG) co-loaded tumor cells with a hydrogel matrix. Activation of PVAX by 808 nm NIR laser irradiation significantly inhibits the tumor relapse by promoting the maturation of dendritic cells and eliciting tumor infiltration of cytotoxic T lymphocytes. A mechanical study reveals that NIR light-triggered antigen release and JQ1-mediated PD-L1 checkpoint blockade cumulatively contribute to the satisfied therapeutic effect. Furthermore, PVAX prepared from the autologous tumor cells induces patient-specific memory immune response to prevent tumor recurrence and metastasis. The PVAX model might provide novel insights for postoperative immunotherapy. Cancer vaccines represent a promising personalized therapeutic approach to treating cancer. Here, the authors report the efficacy in a metastatic model of a cancer vaccine-mediated postoperative immunotherapy, based on the coencapsulation of the JQ1 and a photosensitizer ICG together with inactivated tumor cells into a hydrogel matrix.
Collapse
|
41
|
Leng F, Liu F, Yang Y, Wu Y, Tian W. Strategies on Nanodiagnostics and Nanotherapies of the Three Common Cancers. NANOMATERIALS 2018; 8:nano8040202. [PMID: 29597315 PMCID: PMC5923532 DOI: 10.3390/nano8040202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 03/18/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
The emergence of nanomedicine has enriched the knowledge and strategies of treating diseases, and especially some incurable diseases, such as cancers, acquired immune deficiency syndrome (AIDS), and neurodegenerative diseases. The application of nanoparticles in medicine is in the core of nanomedicine. Nanoparticles can be used in drug delivery for improving the uptake of poorly soluble drugs, targeted delivery to a specific site, and drug bioavailability. Early diagnosis of and targeted therapies for cancers can significantly improve patients' quality of life and extend patients' lives. The advantages of nanoparticles have given them a progressively important role in the nanodiagnosis and nanotherapy of common cancers. To provide a reference for the further application of nanoparticles, this review focuses on the recent development and application of nanoparticles in the early diagnosis and treatment of the three common cancers (lung cancer, liver cancer, and breast cancer) by using quantum dots, magnetic nanoparticles, and gold nanoparticles.
Collapse
Affiliation(s)
- Fan Leng
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Fang Liu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yongtao Yang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Yu Wu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| | - Weiqun Tian
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|