1
|
Nankivell V, Vidanapathirana AK, Hoogendoorn A, Tan JTM, Verjans J, Psaltis PJ, Hutchinson MR, Gibson BC, Lu Y, Goldys E, Zheng G, Bursill CA. Targeting macrophages with multifunctional nanoparticles to detect and prevent atherosclerotic cardiovascular disease. Cardiovasc Res 2024; 120:819-838. [PMID: 38696700 PMCID: PMC11218693 DOI: 10.1093/cvr/cvae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/29/2024] [Accepted: 04/02/2024] [Indexed: 05/04/2024] Open
Abstract
Despite the emergence of novel diagnostic, pharmacological, interventional, and prevention strategies, atherosclerotic cardiovascular disease remains a significant cause of morbidity and mortality. Nanoparticle (NP)-based platforms encompass diverse imaging, delivery, and pharmacological properties that provide novel opportunities for refining diagnostic and therapeutic interventions for atherosclerosis at the cellular and molecular levels. Macrophages play a critical role in atherosclerosis and therefore represent an important disease-related diagnostic and therapeutic target, especially given their inherent ability for passive and active NP uptake. In this review, we discuss an array of inorganic, carbon-based, and lipid-based NPs that provide magnetic, radiographic, and fluorescent imaging capabilities for a range of highly promising research and clinical applications in atherosclerosis. We discuss the design of NPs that target a range of macrophage-related functions such as lipoprotein oxidation, cholesterol efflux, vascular inflammation, and defective efferocytosis. We also provide examples of NP systems that were developed for other pathologies such as cancer and highlight their potential for repurposing in cardiovascular disease. Finally, we discuss the current state of play and the future of theranostic NPs. Whilst this is not without its challenges, the array of multifunctional capabilities that are possible in NP design ensures they will be part of the next frontier of exciting new therapies that simultaneously improve the accuracy of plaque diagnosis and more effectively reduce atherosclerosis with limited side effects.
Collapse
Affiliation(s)
- Victoria Nankivell
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Achini K Vidanapathirana
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Ayla Hoogendoorn
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
| | - Joanne T M Tan
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Johan Verjans
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Peter J Psaltis
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Mark R Hutchinson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| | - Brant C Gibson
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Science, RMIT University, Melbourne, Victoria, Australia
| | - Yiqing Lu
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- School of Engineering, Macquarie University, Sydney, NSW, Australia
| | - Ewa Goldys
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Graduate School of Biomedical Engineering, University of New South Wales, High Street, NSW, 2052, Australia
| | - Gang Zheng
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, M5G 1L7, Canada
| | - Christina A Bursill
- Australian Research Council (ARC) Centre of Excellence for Nanoscale BioPhotonics (CNBP)
- Vascular Research Centre, Lifelong Health, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, 5000, Australia
- Faculty of Health and Medical Science, The University of Adelaide, North Terrace, Adelaide, 5000, Australia
| |
Collapse
|
2
|
Sandbhor P, Palkar P, Bhat S, John G, Goda JS. Nanomedicine as a multimodal therapeutic paradigm against cancer: on the way forward in advancing precision therapy. NANOSCALE 2024. [PMID: 38470224 DOI: 10.1039/d3nr06131k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
Recent years have witnessed dramatic improvements in nanotechnology-based cancer therapeutics, and it continues to evolve from the use of conventional therapies (chemotherapy, surgery, and radiotherapy) to increasingly multi-complex approaches incorporating thermal energy-based tumor ablation (e.g. magnetic hyperthermia and photothermal therapy), dynamic therapy (e.g. photodynamic therapy), gene therapy, sonodynamic therapy (e.g. ultrasound), immunotherapy, and more recently real-time treatment efficacy monitoring (e.g. theranostic MRI-sensitive nanoparticles). Unlike monotherapy, these multimodal therapies (bimodal, i.e., a combination of two therapies, and trimodal, i.e., a combination of more than two therapies) incorporating nanoplatforms have tremendous potential to improve the tumor tissue penetration and retention of therapeutic agents through selective active/passive targeting effects. These combinatorial therapies can correspondingly alleviate drug response against hypoxic/acidic and immunosuppressive tumor microenvironments and promote/induce tumor cell death through various multi-mechanisms such as apoptosis, autophagy, and reactive oxygen-based cytotoxicity, e.g., ferroptosis, etc. These multi-faced approaches such as targeting the tumor vasculature, neoangiogenic vessels, drug-resistant cancer stem cells (CSCs), preventing intra/extravasation to reduce metastatic growth, and modulation of antitumor immune responses work complementary to each other, enhancing treatment efficacy. In this review, we discuss recent advances in different nanotechnology-mediated synergistic/additive combination therapies, emphasizing their underlying mechanisms for improving cancer prognosis and survival outcomes. Additionally, significant challenges such as CSCs, hypoxia, immunosuppression, and distant/local metastasis associated with therapy resistance and tumor recurrences are reviewed. Furthermore, to improve the clinical precision of these multimodal nanoplatforms in cancer treatment, their successful bench-to-clinic translation with controlled and localized drug-release kinetics, maximizing the therapeutic window while addressing safety and regulatory concerns are discussed. As we advance further, exploiting these strategies in clinically more relevant models such as patient-derived xenografts and 3D organoids will pave the way for the application of precision therapy.
Collapse
Affiliation(s)
- Puja Sandbhor
- Institute for NanoBioTechnology, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Pranoti Palkar
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Sakshi Bhat
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Geofrey John
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| | - Jayant S Goda
- Radiobiology, Department of Radiation Oncology & Homi Bhabha National Institute, Mumbai, 400012, India
| |
Collapse
|
3
|
Yasir M, Mishra R, Tripathi AS, Maurya RK, Shahi A, Zaki MEA, Al Hussain SA, Masand VH. Theranostics: a multifaceted approach utilizing nano-biomaterials. DISCOVER NANO 2024; 19:35. [PMID: 38407670 PMCID: PMC10897124 DOI: 10.1186/s11671-024-03979-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/19/2024] [Indexed: 02/27/2024]
Abstract
Biomaterials play a vital role in targeting therapeutics. Over the years, several biomaterials have gained wide attention in the treatment and diagnosis of diseases. Scientists are trying to make more personalized treatments for different diseases, as well as discovering novel single agents that can be used for prognosis, medication administration, and keeping track of how a treatment works. Theranostics based on nano-biomaterials have higher sensitivity and specificity for disease management than conventional techniques. This review provides a concise overview of various biomaterials, including carbon-based materials like fullerenes, graphene, carbon nanotubes (CNTs), and carbon nanofibers, and their involvement in theranostics of different diseases. In addition, the involvement of imaging techniques for theranostics applications was overviewed. Theranostics is an emerging strategy that has great potential for enhancing the accuracy and efficacy of medicinal interventions. Despite the presence of obstacles such as disease heterogeneity, toxicity, reproducibility, uniformity, upscaling production, and regulatory hurdles, the field of medical research and development has great promise due to its ability to provide patients with personalised care, facilitate early identification, and enable focused treatment.
Collapse
Affiliation(s)
- Mohammad Yasir
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector125, Noida, 201313, India.
| | - Ratnakar Mishra
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector125, Noida, 201313, India
| | | | - Rahul K Maurya
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector125, Noida, 201313, India
| | - Ashutosh Shahi
- Amity Institute of Pharmacy, Lucknow, Amity University Uttar Pradesh, Sector125, Noida, 201313, India
| | - Magdi E A Zaki
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 13318, Saudi Arabia.
| | - Sami A Al Hussain
- Department of Chemistry, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, 13318, Saudi Arabia
| | - Vijay H Masand
- Department of Chemistry, Vidya Bharati Mahavidyalaya, Amravati, Maharashtra, India
| |
Collapse
|
4
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
5
|
Han J, Liu Y, Peng D, Liu J, Wu D. Biomedical Application of Porphyrin-Based Amphiphiles and Their Self-Assembled Nanomaterials. Bioconjug Chem 2023; 34:2155-2180. [PMID: 37955349 DOI: 10.1021/acs.bioconjchem.3c00432] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Porphyrins have been vastly explored and applied in many cutting-edge fields with plenty of encouraging achievements because of their excellent properties. As important derivatives of porphyrins, porphyrin-based amphiphiles (PBAs) not only maintain the advanced properties of porphyrins (catalysis, imaging, and energy transfer) but also possess self-assembly and encapsulation capability in aqueous solution. Accordingly, PBAs and their self-assembles have had important roles in diagnosing and treating tumors and inflammation lesions in vivo, but not limited to these. In this article, we introduce the research progress of PBAs, including their constitution, structure design strategies, and performances in tumor and inflammation lesion diagnosis and treatments. On that basis, the defects of synthesized PBAs during their application and the possible effective strategies to overcome the limitations are also proposed. Finally, perspectives on PBAs exploration are updated based on our knowledge. We hope this review will bring researchers from various domains insights about PBAs.
Collapse
Affiliation(s)
- Jialei Han
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Gongchang Road 66, Guangming, Shenzhen, Guangdong 518107, China
| | - Yadong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Gongchang Road 66, Guangming, Shenzhen, Guangdong 518107, China
| | - Danfeng Peng
- Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong 518119, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Gongchang Road 66, Guangming, Shenzhen, Guangdong 518107, China
| | - Dalin Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Gongchang Road 66, Guangming, Shenzhen, Guangdong 518107, China
| |
Collapse
|
6
|
Batool S, Sohail S, Ud Din F, Alamri AH, Alqahtani AS, Alshahrani MA, Alshehri MA, Choi HG. A detailed insight of the tumor targeting using nanocarrier drug delivery system. Drug Deliv 2023; 30:2183815. [PMID: 36866455 DOI: 10.1080/10717544.2023.2183815] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Human struggle against the deadly disease conditions is continued since ages. The contribution of science and technology in fighting against these diseases cannot be ignored exclusively due to the invention of novel procedure and products, extending their size ranges from micro to nano. Recently nanotechnology has been gaining more consideration for its ability to diagnose and treat different cancers. Different nanoparticles have been used to evade the issues related with conservative anticancer delivery systems, including their nonspecificity, adverse effects and burst release. These nanocarriers including, solid lipid nanoparticles (SLNs), liposomes, nano lipid carriers (NLCs), nano micelles, nanocomposites, polymeric and magnetic nanocarriers, have brought revolutions in antitumor drug delivery. Nanocarriers improved the therapeutic efficacy of anticancer drugs with better accumulation at the specific site with sustained release, improved bioavailability and apoptosis of the cancer cells while bypassing the normal cells. In this review, the cancer targeting techniques and surface modification on nanoparticles are discussed briefly with possible challenges and opportunities. It can be concluded that understanding the role of nanomedicine in tumor treatment is significant, and therefore, the modern progressions in this arena is essential to be considered for a prosperous today and an affluent future of tumor patients.
Collapse
Affiliation(s)
- Sibgha Batool
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saba Sohail
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ahmad S Alqahtani
- Department of Pharmacy, Mental Health Hospital, Ministry of Health, Abha, Saudi Arabia
| | - Mohammad A Alshahrani
- Department of Medical Supply in Khamis Mushet General Hospital, Ministry of Health, Khamis Mushet, Saudi Arabia
| | - Mohammed A Alshehri
- Department of Pharmacy, Abha Maternity and Children Hospital, Ministry of Health, Abha, Saudi Arabia
| | - Han Gon Choi
- College of Pharmacy & Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| |
Collapse
|
7
|
Singh AK, Malviya R, Prajapati B, Singh S, Yadav D, Kumar A. Nanotechnology-Aided Advancement in Combating the Cancer Metastasis. Pharmaceuticals (Basel) 2023; 16:899. [PMID: 37375846 PMCID: PMC10304141 DOI: 10.3390/ph16060899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/28/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Modern medicine has been working to find a cure for cancer for almost a century, but thus far, they have not been very successful. Although cancer treatment has come a long way, more work has to be carried out to boost specificity and reduce systemic toxicity. The diagnostic industry is on the cusp of a technological revolution, and early diagnosis is essential for improving prognostic outlook and patient quality of life. In recent years, nanotechnology's use has expanded, demonstrating its efficacy in enhancing fields such as cancer treatment, radiation therapy, diagnostics, and imaging. Applications for nanomaterials are diverse, ranging from enhanced radiation adjuvants to more sensitive early detection instruments. Cancer, particularly when it has spread beyond the original site of cancer, is notoriously tough to combat. Many people die from metastatic cancer, which is why it remains a huge issue. Cancer cells go through a sequence of events known as the "metastatic cascade" throughout metastasis, which may be used to build anti-metastatic therapeutic techniques. Conventional treatments and diagnostics for metastasis have their drawbacks and hurdles that must be overcome. In this contribution, we explore in-depth the potential benefits that nanotechnology-aided methods might offer to the detection and treatment of metastatic illness, either alone or in conjunction with currently available conventional procedures. Anti-metastatic drugs, which can prevent or slow the spread of cancer throughout the body, can be more precisely targeted and developed with the help of nanotechnology. Furthermore, we talk about how nanotechnology is being applied to the treatment of patients with cancer metastases.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Bhupendra Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Deepika Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India; (A.K.S.); (D.Y.)
| | - Arvind Kumar
- Chandigarh Engineering College, Jhanjeri, Mohali 140307, India;
| |
Collapse
|
8
|
Hegde M, Naliyadhara N, Unnikrishnan J, Alqahtani MS, Abbas M, Girisa S, Sethi G, Kunnumakkara AB. Nanoparticles in the diagnosis and treatment of cancer metastases: Current and future perspectives. Cancer Lett 2023; 556:216066. [PMID: 36649823 DOI: 10.1016/j.canlet.2023.216066] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Metastasis accounts for greater than 90% of cancer-related deaths. Despite recent advancements in conventional chemotherapy, immunotherapy, targeted therapy, and their rational combinations, metastatic cancers remain essentially untreatable. The distinct obstacles to treat metastases include their small size, high multiplicity, redundancy, therapeutic resistance, and dissemination to multiple organs. Recent advancements in nanotechnology provide the numerous applications in the diagnosis and prophylaxis of metastatic diseases, including the small particle size to penetrate cell membrane and blood vessels and their capacity to transport complex molecular 'cargo' particles to various metastatic regions such as bones, brain, liver, lungs, and lymph nodes. Indeed, nanoparticles (NPs) have demonstrated a significant ability to target specific cells within these organs. In this regard, the purpose of this review is to summarize the present state of nanotechnology in terms of its application in the diagnosis and treatment of metastatic cancer. We intensively reviewed applications of NPs in fluorescent imaging, PET scanning, MRI, and photoacoustic imaging to detect metastasis in various cancer models. The use of targeted NPs for cancer ablation in conjunction with chemotherapy, photothermal treatment, immuno therapy, and combination therapy is thoroughly discussed. The current review also highlights the research opportunities and challenges of leveraging engineering technologies with cancer cell biology and pharmacology to fabricate nanoscience-based tools for treating metastases.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nikunj Naliyadhara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Jyothsna Unnikrishnan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, 61421, Saudi Arabia; BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, LE1 7RH, UK
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, 61421, Saudi Arabia; Computers and Communications Department, College of Engineering, Delta University for Science and Technology, Gamasa, 35712, Egypt
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
9
|
Liao M, Cui J, Yang M, Wei Z, Xie Y, Lu C. Photoinduced electron transfer in metalloporphyrins. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
10
|
Avula LR, Grodzinski P. Nanotechnology-aided advancement in the combating of cancer metastasis. Cancer Metastasis Rev 2022; 41:383-404. [PMID: 35366154 PMCID: PMC8975728 DOI: 10.1007/s10555-022-10025-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/09/2022] [Indexed: 02/03/2023]
Abstract
Cancer, especially when it has metastasized to different locations in the body, is notoriously difficult to treat. Metastatic cancer accounts for most cancer deaths and thus remains an enormous challenge. During the metastasis process, cancer cells negotiate a series of steps termed the “metastatic cascadeˮ that offer potential for developing anti-metastatic therapy strategies. Currently available conventional treatment and diagnostic methods addressing metastasis come with their own pitfalls and roadblocks. In this contribution, we comprehensively discuss the potential improvements that nanotechnology-aided approaches are able to bring, either alone or in combination with the existing conventional techniques, to the identification and treatment of metastatic disease. We tie specific nanotechnology-aided strategies to the complex biology of the different steps of the metastatic cascade in order to open up new avenues for fine-tuned targeting and development of anti-metastatic agents designed specifically to prevent or mitigate the metastatic outgrowth of cancer. We also present a viewpoint on the progress of translation of nanotechnology into cancer metastasis patient care.
Collapse
Affiliation(s)
- Leela Rani Avula
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| | - Piotr Grodzinski
- Nanodelivery Systems and Devices Branch, Cancer Imaging Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
11
|
Oxygen saturation and blood volume analysis by Photoacoustic imaging to identify pre and post-PDT vascular changes. Saudi J Biol Sci 2022; 29:103304. [PMID: 35574285 PMCID: PMC9092990 DOI: 10.1016/j.sjbs.2022.103304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/07/2022] [Accepted: 04/17/2022] [Indexed: 11/22/2022] Open
Abstract
In this study, the blood volume and oxygen saturation of tumors were measured after photoacoustic imaging (PAI) under conditions of pre-photodynamic therapy (PDT), post-PDT, and 4 hrs, and 24 hrs post-PDT. PDTs with aminolevulinic acid (ALA) and low and high doses of benzoporphyrin derivative (BPD) were conducted to observe oxygen saturation changes, and the rapid oxygen consumption in the blood detected due to the action of BPD at the vascular level resulted in the recovery of PDT completion. Likewise, blood volume changes followed by ALA-PDT and BPD-PDT at low and high doses depicted a fast expansion of the blood volume after treatment. The tumor subjected to a high dose of ALA-PDT showed a partial alteration of Hb-pO2 in the first 24 hrs, as did the tumors treated with two ALA- and BPD-mediated PDTs. The Hb-pO2 started reducing immediately post-PDT and was less than 30% after 4 hrs until 24 hrs post-PDT. Reduced vascular demand was possibly due to tumor necrosis, as shown by the permanent damage in the cancer cells' bioluminescence signal. The ALA-mediated PDT-subjected tumor showed a 50% drop in BV at 24 hrs post-PDT, which is suggestive of vascular pruning. The studied data of blood volume against BLI showed the blood volume and oxygenation variations validating the cells' metabolic activity, including cell death.
Collapse
|
12
|
Qindeel M, Sargazi S, Hosseinikhah SM, Rahdar A, Barani M, Thakur VK, Pandey S, Mirsafaei R. Porphyrin‐Based Nanostructures for Cancer Theranostics: Chemistry, Fundamentals and Recent Advances. ChemistrySelect 2021. [DOI: 10.1002/slct.202103418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Maimoona Qindeel
- Hamdard Institute of Pharmaceutical Sciences Hamdard University Islamabad Campus Islamabad Pakistan
- Department of Pharmacy Quaid-i-Azam University Islamabad Pakistan
| | - Saman Sargazi
- Cellular and Molecular Research Center Research Institute of Cellular and Molecular Sciences in Infectious Diseases Zahedan University of Medical Sciences Zahedan 9816743463 Iran
| | - Seyedeh Maryam Hosseinikhah
- Nanotechnology Research Center Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Abbas Rahdar
- Department of Physics Faculty of Science University of Zabol Zabol Iran
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research Center Kerman University of Medical Sciences Kerman 7616913555 Iran
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre Scotland's Rural College Scotland Edinburgh EH9 3JG United Kingdom
- School of Engineering University of Petroleum & Energy Studies (UPES) Dehradun 248007 Uttarakhand India
| | - Sadanand Pandey
- Particulate Matter Research Center Research Institute of Industrial Science & Technology (RIST) 187-12, Geumho-ro Gwangyang-si Jeollanam-do 57801, Republic of Korea
| | - Razieh Mirsafaei
- Novel Drug Delivery Systems Research Centre and Department of Pharmaceutics School of Pharmacy Isfahan University of Medical Sciences Isfahan Iran
| |
Collapse
|
13
|
Bukhari SZ, Zeth K, Iftikhar M, Rehman M, Usman Munir M, Khan WS, Ihsan A. Supramolecular lipid nanoparticles as delivery carriers for non-invasive cancer theranostics. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100067. [PMID: 34909685 PMCID: PMC8663983 DOI: 10.1016/j.crphar.2021.100067] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotheranostics is an emerging frontier of personalized medicine research particularly for cancer, which is the second leading cause of death. Supramolecular aspects in theranostics are quite allured to achieve more regulation and controlled features. Supramolecular nanotheranostics architecture is focused on engineering of modular supramolecular assemblies benefitting from their mutable and stimuli-responsive properties which confer an ultimate potential for the fabrication of unified innovative nanomedicines with controlled features. Amalgamation of supramolecular approaches to nano-based features further equip the potential of designing novel approaches to overcome limitations seen by the conventional theranostic strategies, for curing even the lethal diseases and endowing personalized therapeutics with optimistic prognosis, endorsing their clinical translation. Among many potential nanocarriers for theranostics, lipid nanoparticles (LNPs) have shown various promising advances in theranostics and their formulation can be tailored for several applications. Despite the great advancement in cancer nanotheranostics, there are still many challenges that need to be highlighted to fill the literature gap. For this purpose, herein, we have presented a systematic overview on the subject and proposed LNPs as the potential material to manage cancer via non-invasive approaches by highlighting the use of supramolecular approaches to make them robust for cancer theranostics. We have concluded the review by entailing the future perspectives of lipid nanotheranostics towards clinical translation.
Collapse
Affiliation(s)
- Syeda Zunaira Bukhari
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Kornelius Zeth
- Department of Science and Environment, Roskilde University Center, DK-4000 Roskilde, Denmark
| | - Maryam Iftikhar
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Mubashar Rehman
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Muhammad Usman Munir
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 72388, Saudi Arabia
| | - Waheed S. Khan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Ayesha Ihsan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| |
Collapse
|
14
|
Ackun-Farmmer MA, Overby CT, Haws BE, Choe R, Benoit DSW. Biomaterials for Orthopaedic Diagnostics and Theranostics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 19. [PMID: 34458652 DOI: 10.1016/j.cobme.2021.100308] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Despite widespread use of conventional diagnostic methods in orthopaedic applications, limitations still exist in detection and diagnosing many pathologies especially at early stages when intervention is most critical. The use of biomaterials to develop diagnostics and theranostics, including nanoparticles and scaffolds for systemic or local applications, has significant promise to address these shortcomings and enable successful clinical translation. These developments in both modular and holistic design of diagnostic and theranostic biomaterials may improve patient treatments for myriad orthopaedic applications ranging from cancer to fractures to infection.
Collapse
Affiliation(s)
- Marian A Ackun-Farmmer
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Clyde T Overby
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| | - Brittany E Haws
- Department of Orthopaedics, University of Rochester, Rochester, NY, USA
| | - Regine Choe
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Department of Electrical and Computer Engineering, University of Rochester, Rochester, NY, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA.,Department of Orthopaedics, University of Rochester, Rochester, NY, USA.,Materials Science Program, University of Rochester, Rochester, NY, USA.,Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
15
|
Chang E, Bu J, Ding L, Lou JWH, Valic MS, Cheng MHY, Rosilio V, Chen J, Zheng G. Porphyrin-lipid stabilized paclitaxel nanoemulsion for combined photodynamic therapy and chemotherapy. J Nanobiotechnology 2021; 19:154. [PMID: 34034749 PMCID: PMC8147067 DOI: 10.1186/s12951-021-00898-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/15/2021] [Indexed: 01/09/2023] Open
Abstract
Background Porphyrin-lipids are versatile building blocks that enable cancer theranostics and have been applied to create several multimodal nanoparticle platforms, including liposome-like porphysome (aqueous-core), porphyrin nanodroplet (liquefied gas-core), and ultrasmall porphyrin lipoproteins. Here, we used porphyrin-lipid to stabilize the water/oil interface to create porphyrin-lipid nanoemulsions with paclitaxel loaded in the oil core (PLNE-PTX), facilitating combination photodynamic therapy (PDT) and chemotherapy in one platform. Results PTX (3.1 wt%) and porphyrin (18.3 wt%) were loaded efficiently into PLNE-PTX, forming spherical core–shell nanoemulsions with a diameter of 120 nm. PLNE-PTX demonstrated stability in systemic delivery, resulting in high tumor accumulation (~ 5.4 ID %/g) in KB-tumor bearing mice. PLNE-PTX combination therapy inhibited tumor growth (78%) in an additive manner, compared with monotherapy PDT (44%) or chemotherapy (46%) 16 days post-treatment. Furthermore, a fourfold reduced PTX dose (1.8 mg PTX/kg) in PLNE-PTX combination therapy platform demonstrated superior therapeutic efficacy to Taxol at a dose of 7.2 mg PTX/kg, which can reduce side effects. Moreover, the intrinsic fluorescence of PLNE-PTX enabled real-time tracking of nanoparticles to the tumor, which can help inform treatment planning. Conclusion PLNE-PTX combining PDT and chemotherapy in a single platform enables superior anti-tumor effects and holds potential to reduce side effects associated with monotherapy chemotherapy. The inherent imaging modality of PLNE-PTX enables real-time tracking and permits spatial and temporal regulation to improve cancer treatment. Graphic Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-00898-1.
Collapse
Affiliation(s)
- Enling Chang
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada.,Institute of Biomedical Engineering, University of Toronto, PMCRT 5-354, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Jiachuan Bu
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Lili Ding
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Jenny W H Lou
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael S Valic
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada.,Institute of Biomedical Engineering, University of Toronto, PMCRT 5-354, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Miffy H Y Cheng
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada
| | - Véronique Rosilio
- Institut Galien Paris-Saclay, Université Paris-Saclay, CNRS, Châtenay-Malabry, France
| | - Juan Chen
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, PMCRT 5-353, 101 College Street, Toronto, ON, M5G 1L7, Canada. .,Institute of Biomedical Engineering, University of Toronto, PMCRT 5-354, 101 College Street, Toronto, ON, M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Wang J, Huang J, Zhou W, Zhao J, Peng Q, Zhang L, Wang Z, Li P, Li R. Hypoxia modulation by dual-drug nanoparticles for enhanced synergistic sonodynamic and starvation therapy. J Nanobiotechnology 2021; 19:87. [PMID: 33771168 PMCID: PMC7995598 DOI: 10.1186/s12951-021-00837-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Sonodynamic therapy (SDT) is an emerging non-invasive therapeutic technique. SDT-based cancer therapy strategies are presently underway, and it may be perceived as a promising approach to improve the efficiency of anti-cancer treatment. In this work, multifunctional theranostic nanoparticles (NPs) were synthesized for synergistic starvation therapy and SDT by loading glucose oxidase (GOx, termed G) and 5,10,15,20-tetrakis (4-chlorophenyl) porphyrin) Cl (T (p-Cl) PPMnCl, termed PMnC) in Poly (lactic-co-glycolic) acid (PLGA) NPs (designated as MG@P NPs). RESULTS On account of the peroxidase-like activity of PMnC, MG@P NPs can catalyze hydrogen peroxide (H2O2) in tumor regions to produce oxygen (O2), thus enhancing synergistic therapeutic effects by accelerating the decomposition of glucose and promoting the production of cytotoxic singlet oxygen (1O2) induced by ultrasound (US) irradiation. Furthermore, the NPs can also serve as excellent photoacoustic (PA)/magnetic resonance (MR) imaging contrast agents, effectuating imaging-guided cancer treatment. CONCLUSION Multifunctional MG@P NPs can effectuate the synergistic amplification effect of cancer starvation therapy and SDT by hypoxia modulation, and act as contrast agents to enhance MR/PA dual-modal imaging. Consequently, MG@P NPs might be a promising nano-platform for highly efficient cancer theranostics.
Collapse
Affiliation(s)
- Jingxue Wang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Ju Huang
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Weichen Zhou
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Jiawen Zhao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Qi Peng
- University-Town Hospital, Chongqing Medical University, Chongqing, 401331, People's Republic of China
| | - Liang Zhang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Pan Li
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, Institute of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Rui Li
- Department of Ultrasound, The Third Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
17
|
Li AC, Ong YH, Li C, He J, Dimofte A, Busch TM, Wilson BC, Weersink R, Zhu TC. A Comparison of Two Probes to Determine Rectum Optical Properties. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2021; 11628:1162808. [PMID: 34083859 PMCID: PMC8171236 DOI: 10.1117/12.2582395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Tissue optical properties are crucial for determining the light dose delivered to the tumor. Two probes are compared: the two-catheter probe is based on transmittance measurement between one point source and one isotropic detector inside parallel catheters spaced at 0.5 cm along a 1-inch diameter transparent cylinder; and a 1-inch trans-rectal diffuse optical tomography (DOT) probe designed for prostate measurements, using a multiple fiber-array with source-detector separations of 1.4-10 mm. The two-catheter probe uses an empirical model for primary and scatter light fluence rates in the cylindrical cavity condition for anal PDT to determine optical properties along the source catheter using dual motors to move the source and detector along the catheters. The DOT probe uses finite element method (FEM) to obtain distribution of optical properties in 3D. Validations for the two probes were performed in liquid and solid phantoms. For each method, validation was performed in tissue-mimicking liquid phantoms for a range of known optical properties (μa between 0.05 and 0.9 cm-1 and μs' between 5.5 and 16.5 cm-1). To cross-check the two methods, solid phantoms were created of known optical properties at the University of Pennsylvania and sent for measurement to Princess Margaret Cancer Centre (PMH) to mimic realistic patient simulating conditions. Measurements were taken and optical properties were then recovered without knowing the expected values to cross-validate each probe. The results show modest agreement between the measured μa and μs'values, but high degree of agreement between the measured μeff performed independently using the two methods.
Collapse
Affiliation(s)
- Andrew C Li
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104
| | - Yi Hong Ong
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104
| | - Celina Li
- University of Toronto/University Health Network, Toronto, ON M5G 2C4, Canada
| | - Jie He
- University of Toronto/University Health Network, Toronto, ON M5G 2C4, Canada
| | - Andreea Dimofte
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104
| | - Theresa M Busch
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104
| | - Brian C Wilson
- University of Toronto/University Health Network, Toronto, ON M5G 2C4, Canada
| | - Robert Weersink
- University of Toronto/University Health Network, Toronto, ON M5G 2C4, Canada
| | - Timothy C Zhu
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
18
|
Yu J, Liu Y, Zhou S, Wang Y, Wang Y. Stimuli-responsive phospholipid-drug conjugates (PDCs)-based nanovesicles for drug delivery and theranostics. Int J Pharm 2020; 590:119920. [PMID: 33002539 DOI: 10.1016/j.ijpharm.2020.119920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 01/07/2023]
Abstract
Liposomes represent one of the most successful nano-drug delivery systems among enormous nano-carriers. Although great progress has been made in conventional liposomes, the emerging shortcomings still impair the therapeutic index. The proposal of stimuli-responsive phospholipid-drug conjugates (PDCs)-based nanovesicles solves the challenges that conventional liposomes are faced with, showing great potential for cancer diagnosis and therapy. Herein, we intend to overview the current progress and unique advantages of stimuli-responsive PDCs-based nanovesicles. First, the challenges of conventional liposomes and the development of PDCs-based nanovesicles are summarized. Next, the stimuli-responsive elements used in current stimuli-responsive PDCs-based nanovesicles are outlined. Then, the unique superiorities of stimuli-responsive PDCs-based nanovesicles for drug delivery and theranostics are highlighted in detail. Finally, the future opportunities and challenges of stimuli-responsive PDCs-based nanovesicles for clinical translation are put forward.
Collapse
Affiliation(s)
- Jiang Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ying Liu
- National Institute for Food and Drug Control, Beijing 102629, China
| | - Shuang Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yingli Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China.
| |
Collapse
|
19
|
Simões JCS, Sarpaki S, Papadimitroulas P, Therrien B, Loudos G. Conjugated Photosensitizers for Imaging and PDT in Cancer Research. J Med Chem 2020; 63:14119-14150. [PMID: 32990442 DOI: 10.1021/acs.jmedchem.0c00047] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Early cancer detection and perfect understanding of the disease are imperative toward efficient treatments. It is straightforward that, for choosing a specific cancer treatment methodology, diagnostic agents undertake a critical role. Imaging is an extremely intriguing tool since it assumes a follow up to treatments to survey the accomplishment of the treatment and to recognize any conceivable repeating injuries. It also permits analysis of the disease, as well as to pursue treatment and monitor the possible changes that happen on the tumor. Likewise, it allows screening the adequacy of treatment and visualizing the state of the tumor. Additionally, when the treatment is finished, observing the patient is imperative to evaluate the treatment methodology and adjust the treatment if necessary. The goal of this review is to present an overview of conjugated photosensitizers for imaging and therapy.
Collapse
Affiliation(s)
- João C S Simões
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland.,BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | - Sophia Sarpaki
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| | | | - Bruno Therrien
- Institute of Chemistry, University of Neuchatel, Avenue de Bellevaux 51, CH-2000 Neuchatel, Switzerland
| | - George Loudos
- BioEmission Technology Solutions, Alexandras Avenue 116, 11472 Athens, Greece
| |
Collapse
|
20
|
Fathi P, Pan D. Current trends in pyrrole and porphyrin-derived nanoscale materials for biomedical applications. Nanomedicine (Lond) 2020; 15:2493-2515. [PMID: 32975469 PMCID: PMC7610151 DOI: 10.2217/nnm-2020-0125] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/14/2020] [Indexed: 02/01/2023] Open
Abstract
This article is written to provide an up-to-date review of pyrrole-based biomedical materials. Porphyrins and other tetrapyrrolic molecules possess unique magnetic, optical and other photophysical properties that make them useful for bioimaging and therapy. This review touches briefly on some of the synthetic strategies to obtain porphyrin- and tetrapyrrole-based nanoparticles, as well as the variety of applications in which crosslinked, self-assembled, porphyrin-coated and other nanoparticles are utilized. We explore examples of these nanoparticles' applications in photothermal therapy, drug delivery, photodynamic therapy, stimuli response, fluorescence imaging, photoacoustic imaging, magnetic resonance imaging, computed tomography and positron emission tomography. We anticipate that this review will provide a comprehensive summary of pyrrole-derived nanoparticles and provide a guideline for their further development.
Collapse
Affiliation(s)
- Parinaz Fathi
- Departments of Bioengineering, Materials Science & Engineering & Beckman Institute, University of Illinois, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
| | - Dipanjan Pan
- Departments of Bioengineering, Materials Science & Engineering & Beckman Institute, University of Illinois, Urbana, IL 61801, USA
- Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL 61801, USA
- Departments of Diagnostic Radiology & Nuclear Medicine & Pediatrics, University of Maryland Baltimore, Health Sciences Facility III, 670 W Baltimore St., Baltimore, MD 21201, USA
- Department of Chemical, Biochemical & Environmental Engineering, University of Maryland Baltimore County, Interdisciplinary Health Sciences Facility, 1000 Hilltop Circle Baltimore, MD 21250, USA
| |
Collapse
|
21
|
Gao X, Li L, Cai X, Huang Q, Xiao J, Cheng Y. Targeting nanoparticles for diagnosis and therapy of bone tumors: Opportunities and challenges. Biomaterials 2020; 265:120404. [PMID: 32987273 DOI: 10.1016/j.biomaterials.2020.120404] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022]
Abstract
A variety of targeted nanoparticles were developed for the diagnosis and therapy of orthotopic and metastatic bone tumors during the past decade. This critical review will focus on principles and methods in the design of these bone-targeted nanoparticles. Ligands including bisphosphonates, aspartic acid-rich peptides and synthetic polymers were grafted on nanoparticles such as PLGA nanoparticles, liposomes, dendrimers and inorganic nanoparticles for bone targeting. Besides, other ligands such as monoclonal antibodies, peptides and aptamers targeting biomarkers on tumor/bone cells were identified for targeted diagnosis and therapy. Examples of targeted nanoparticles for the early detection of bone metastatic tumors and the ablation of cancer via chemotherapy, photothermal therapy, gene therapy and combination therapy will be intensively reviewed. The development of multifunctional nanoparticles to break down the "vicious" cycle between tumor cell proliferation and bone resorption, and the challenges and perspectives in this area will be discussed.
Collapse
Affiliation(s)
- Xin Gao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Lin Li
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Xiaopan Cai
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Quan Huang
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Jianru Xiao
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Department of Orthopedics Oncology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China.
| | - Yiyun Cheng
- East China Normal University and Shanghai Changzheng Hospital Joint Research Center for Orthopedic Oncology, 200241, Shanghai, China; Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
22
|
Das SS, Alkahtani S, Bharadwaj P, Ansari MT, ALKahtani MDF, Pang Z, Hasnain MS, Nayak AK, Aminabhavi TM. Molecular insights and novel approaches for targeting tumor metastasis. Int J Pharm 2020; 585:119556. [PMID: 32574684 DOI: 10.1016/j.ijpharm.2020.119556] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 06/01/2020] [Accepted: 06/14/2020] [Indexed: 12/18/2022]
Abstract
In recent years, due to the effective drug delivery and preciseness of tumor sites or microenvironment, the targeted drug delivery approaches have gained ample attention for tumor metastasis therapy. The conventional treatment approaches for metastasis therapy have reported with immense adverse effects because they exhibited maximum probability of killing the carcinogenic cells along with healthy cells. The tumor vasculature, comprising of vasculogenic impressions and angiogenesis, greatly depends upon the growth and metastasis in the tumors. Therefore, various nanocarriers-based delivery approaches for targeting to tumor vasculature have been attempted as efficient and potential approaches for the treatment of tumor metastasis and the associated lesions. Furthermore, the targeted drug delivery approaches have found to be most apt way to overcome from all the limitations and adverse effects associated with the conventional therapies. In this review, various approaches for efficient targeting of pharmacologically active chemotherapeutics against tumor metastasis with the cohesive objectives of prognosis, tracking and therapy are summarized.
Collapse
Affiliation(s)
- Sabya Sachi Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi 835 215, Jharkhand, India
| | - Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Priyanshu Bharadwaj
- UFR des Sciences de Santé, Université de Bourgogne Franche-Comté, Dijon 21000, France
| | - Mohammed Tahir Ansari
- School of Pharmacy, University of Nottingham Malaysia, Jalan Broga, Semenyih, Kajang, Selangor 43500, Malaysia
| | - Muneera D F ALKahtani
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 102275, Riyadh 11675, Saudi Arabia
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai 201203, China
| | - Md Saquib Hasnain
- Department of Pharmacy, Shri Venkateshwara University, NH-24, Rajabpur, Gajraula, Amroha 244236, U.P., India.
| | - Amit Kumar Nayak
- Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj 757086, Odisha, India.
| | | |
Collapse
|
23
|
Liu Y, Wang H, Li S, Chen C, Xu L, Huang P, Liu F, Su Y, Qi M, Yu C, Zhou Y. In situ supramolecular polymerization-enhanced self-assembly of polymer vesicles for highly efficient photothermal therapy. Nat Commun 2020; 11:1724. [PMID: 32265490 PMCID: PMC7138818 DOI: 10.1038/s41467-020-15427-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/09/2020] [Indexed: 11/18/2022] Open
Abstract
Vesicular photothermal therapy agents (PTAs) are highly desirable in photothermal therapy (PTT) for their excellent light-harvesting ability and versatile hollow compartments. However, up to now, the reported vesicular PTAs are generally self-assembled from small molecules like liposomes, and polymer vesicles have seldom been used as PTAs due to the unsatisfactory photothermal conversion efficiency resulting from the irregular packing of chromophores in the vesicle membranes. Here we report a nano-sized polymer vesicle from hyperbranched polyporphyrins with favorable photothermal stability and extraordinarily high photothermal efficiency (44.1%), showing great potential in imaging-guided PTT for tumors through in vitro and in vivo experiments. These excellent properties are attributed to the in situ supramolecular polymerization of porphyrin units inside the vesicle membrane into well-organized 1D monofilaments driven by π–π stacking. We believe the supramolecular polymerization-enhanced self-assembly process reported here will shed a new light on the design of supramolecular materials with new structures and functions. Photothermal therapy (PTT) has recently emerged as a promising approach for cancer therapy. Here, the authors report a hyperbranched polymer vesicle with favorable photothermal stability and high photothermal efficiency for PTT through a supramolecular polymerization-enhanced self-assembly strategy.
Collapse
Affiliation(s)
- Yannan Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Hao Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education, Shenzhen University, 518060, Shenzhen, P. R. China
| | - Shanlong Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Chuanshuang Chen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Li Xu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China.,Joint Research Center for Precision Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, 6600th Nanfeng Road, Fengxian District, 201499, Shanghai, P. R. China
| | - Ping Huang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China.,Joint Research Center for Precision Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital South Campus, 6600th Nanfeng Road, Fengxian District, 201499, Shanghai, P. R. China
| | - Feng Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Yue Su
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Meiwei Qi
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Chunyang Yu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China
| | - Yongfeng Zhou
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, 200240, Shanghai, P. R. China.
| |
Collapse
|
24
|
Zeng Q, Guo Q, Yuan Y, Zhang X, Jiang W, Xiao S, Zhang B, Lou X, Ye C, Liu M, Bouchard LS, Zhou X. A Small Molecular Multifunctional Tool for pH Detection, Fluorescence Imaging, and Photodynamic Therapy. ACS APPLIED BIO MATERIALS 2020; 3:1779-1786. [PMID: 35021667 DOI: 10.1021/acsabm.9b01080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A smart multitool platform for theranostics would be useful for monitoring the administration of therapies in vivo. However, the integration of multiple functions into a single small-molecule platform remains a challenge. In this study, we developed a multifunctional probe based on a small-molecule platform. The properties of this probe were investigated via hyperpolarized 129Xe NMR/MRI, fluorescence imaging in cells and in vivo, and photodynamic therapy (PDT) in tumor mouse models. This multifunctional probe shows good pH response across a broad range of pH values. It also exhibits excellent fluorescence in vivo for mapping its biodistribution. Additionally, it produces enough 1O2 radicals for in vivo PDT. The combination of these functionalities into a single small-molecule platform, rather than a bulky nanoconstruct, offers unique possibilities for molecular imaging and therapy.
Collapse
Affiliation(s)
- Qingbin Zeng
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China
| | - Qianni Guo
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Yaping Yuan
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Xiaoxiao Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China
| | - Weiping Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Sa Xiao
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Bin Zhang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China
| | - Xin Lou
- Department of Radiology, Chinese PLA General Hospital, Beijing, P. R. China
| | - Chaohui Ye
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| | - Louis-S Bouchard
- California Nano Systems Institute, Jonsson Comprehensive Cancer Center, The Molecular Biology Institute, Department of Chemistry and of Bioengineering, University of California, Los Angeles, California, United States
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan 430071, P. R. China.,University of Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
25
|
Gao D, Guo X, Zhang X, Chen S, Wang Y, Chen T, Huang G, Gao Y, Tian Z, Yang Z. Multifunctional phototheranostic nanomedicine for cancer imaging and treatment. Mater Today Bio 2020; 5:100035. [PMID: 32211603 PMCID: PMC7083767 DOI: 10.1016/j.mtbio.2019.100035] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer, as one of the most life-threatening diseases, shows a high fatality rate around the world. When improving the therapeutic efficacy of conventional cancer treatments, researchers also conduct extensive studies into alternative therapeutic approaches, which are safe, valid, and economical. Phototherapies, including photodynamic therapy (PDT) and photothermal therapy (PTT), are tumor-ablative and function-reserving oncologic interventions, showing strong potential in clinical cancer treatment. During phototherapies, the non-toxic phototherapeutic agents can be activated upon light irradiation to induce cell death without causing much damage to normal tissues. Besides, with the rapid development of nanotechnology in the past decades, phototheranostic nanomedicine also has attracted tremendous interests aiming to continuously refine their performance. Herein, we reviewed the recent progress of phototheranostic nanomedicine for improved cancer therapy. After a brief introduction of the therapeutic principles and related phototherapeutic agents for PDT and PTT, the existing works on developing of phototheranostic nanomedicine by mainly focusing on their categories and applications, particularly on phototherapy-synergized cancer immunotherapy, are comprehensively reviewed. More importantly, a brief conclusion and future challenges of phototheranostic nanomedicine from our point of view are delivered in the last part of this article.
Collapse
Affiliation(s)
- D. Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - X. Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - X. Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - S. Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Y. Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - T. Chen
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - G. Huang
- State Key Laboratory of Non-food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, 530007, China
| | - Y. Gao
- Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Number 7 Weiwu Road, Zhengzhou, 450003, China
| | - Z. Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Z. Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
26
|
Assessing micrometastases as a target for nanoparticles using 3D microscopy and machine learning. Proc Natl Acad Sci U S A 2019; 116:14937-14946. [PMID: 31285340 DOI: 10.1073/pnas.1907646116] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Metastasis of solid tumors is a key determinant of cancer patient survival. Targeting micrometastases using nanoparticles could offer a way to stop metastatic tumor growth before it causes excessive patient morbidity. However, nanoparticle delivery to micrometastases is difficult to investigate because micrometastases are small in size and lie deep within tissues. Here, we developed an imaging and image analysis workflow to analyze nanoparticle-cell interactions in metastatic tumors. This technique combines tissue clearing and 3D microscopy with machine learning-based image analysis to assess the physiology of micrometastases with single-cell resolution and quantify the delivery of nanoparticles within them. We show that nanoparticles access a higher proportion of cells in micrometastases (50% nanoparticle-positive cells) compared with primary tumors (17% nanoparticle-positive cells) because they reside close to blood vessels and require a small diffusion distance to reach all tumor cells. Furthermore, the high-throughput nature of our image analysis workflow allowed us to profile the physiology and nanoparticle delivery of 1,301 micrometastases. This enabled us to use machine learning-based modeling to predict nanoparticle delivery to individual micrometastases based on their physiology. Our imaging method allows researchers to measure nanoparticle delivery to micrometastases and highlights an opportunity to target micrometastases with nanoparticles. The development of models to predict nanoparticle delivery based on micrometastasis physiology could enable personalized treatments based on the specific physiology of a patient's micrometastases.
Collapse
|
27
|
Harmatys KM, Overchuk M, Zheng G. Rational Design of Photosynthesis-Inspired Nanomedicines. Acc Chem Res 2019; 52:1265-1274. [PMID: 31021599 DOI: 10.1021/acs.accounts.9b00104] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The sun is the most abundant source of energy on earth. Phototrophs have discovered clever strategies to harvest this light energy and convert it to chemical energy for biomass production. This is achieved in light-harvesting complexes, or antennas, that funnel the exciton energy into the reaction centers. Antennas contain an array of chlorophylls, linear tetrapyrroles, and carotenoid pigments spatially controlled by neighboring proteins. This fine-tuned regulation of protein-pigment arrangements is crucial for survival in the conditions of both excess and extreme light deficit. Photomedicine and photodiagnosis have long been utilizing naturally derived and synthetic monomer dyes for imaging, photodynamic and photothermal therapy; however, the precise regulation of damage inflicted by these therapies requires more complex architectures. In this Account, we discuss how two mechanisms found in photosynthetic systems, photoprotection and light harvesting, have inspired scientists to create nanomedicines for more effective and precise phototherapies. Researchers have been recapitulating natural photoprotection mechanisms by utilizing carotenoids and other quencher molecules toward the design of photodynamic molecular beacons (PDT beacons) for disease-specific photoactivation. We highlight the seminal studies describing peptide-linked porphyrin-carotenoid PDT beacons, which are locally activated by a disease-specific enzyme. Examples of more advanced constructs include tumor-specific mRNA-activatable and polyionic cell-penetrating PDT beacons. An alternative approach toward harnessing photosynthetic processes for biomedical applications includes the design of various nanostructures. This Account will primarily focus on organic lipid-based micro- and nanoparticles. The phenomenon of nonphotochemical quenching, or excess energy release in the form of heat, has been widely explored in the context of porphyrin-containing nanomedicines. These quenched nanostructures can be implemented toward photoacoustic imaging and photothermal therapy. Upon nanostructure disruption, as a result of tissue accumulation and subsequent cell uptake, activatable fluorescence imaging and photodynamic therapy can be achieved. Alternatively, processes found in nature for light harvesting under dim conditions, such as in the deep sea, can be harnessed to maximize light absorption within the tissue. Specifically, high-ordered dye aggregation that results in a bathochromic shift and increased absorption has been exploited for the collection of more light with longer wavelengths, characterized by maximum tissue penetration. Overall, the profound understanding of photosynthetic systems combined with rapid development of nanotechnology has yielded a unique field of nature-inspired photomedicine, which holds promise toward more precise and effective phototherapies.
Collapse
Affiliation(s)
- Kara M. Harmatys
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Marta Overchuk
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Gang Zheng
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Department of Medical Biophysics, University of Toronto, 101 College Street, Toronto, Ontario M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
28
|
Abstract
As unique molecules with both therapeutic and diagnostic properties, porphyrin derivatives have been extensively employed for cancer treatment. Porphyrins not only show powerful phototherapeutic effects (photodynamic and photothermal therapies), but also exhibit excellent imaging capacities, such as near-infrared fluorescent imaging (NIRFI), magnetic resonance imaging (MRI), photoacoustic imaging (PAI), positron emission tomography (PET), and single-photon emission computed tomography (SPECT). In order to take advantage of their robust phototherapeutic effects and excellent imaging capacities, porphyrins can be used to create nanomedicines with effective therapeutic and precise diagnostic properties for cancer treatment. In this Review, we summarize porphyrin-based nanomedicines which have been developed recently, including porphyrin-based liposomes, micelles, polymeric nanoparticles, peptide nanoparticles, and small-molecule nanoassemblies, and their applications on cancer therapy and diagnosis. The outlook and limitation of porphyrin-based nanomedicines are also reviewed.
Collapse
Affiliation(s)
- Xiangdong Xue
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center , University of California Davis , Sacramento , California 95817 , United States
| | - Aaron Lindstrom
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center , University of California Davis , Sacramento , California 95817 , United States
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center , University of California Davis , Sacramento , California 95817 , United States
| |
Collapse
|
29
|
Philp L, Chan H, Rouzbahman M, Overchuk M, Chen J, Zheng G, Bernardini MQ. Use of Porphysomes to detect primary tumour, lymph node metastases, intra-abdominal metastases and as a tool for image-guided lymphadenectomy: proof of concept in endometrial cancer. Am J Cancer Res 2019; 9:2727-2738. [PMID: 31131064 PMCID: PMC6525988 DOI: 10.7150/thno.31225] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022] Open
Abstract
Objective: To investigate Porphysome fluorescence image-guided resection (PYRO-FGR) for detection of uterine tumour, metastatic lymph nodes and abdominal metastases in a model of endometrial cancer. Methods: White New Zealand rabbits were inoculated with VX2 cells via intra-myometrial injection. At 30 days, Porphysomes were administered intravenously. At 24 h the abdomen was imaged and fluorescent tissue identified (PYRO-FGR). After complete resection of fluorescent tissue, fluorescence-negative lymph nodes and peritoneal biopsies were removed. Histopathology including ultra-staging and analysis by a pathologist was used to detect tumour. Fluorescence signal to background ratio (SBR) was calculated and VX2 (+) tissue compared to VX2 (-) tissue. Biodistribution was calculated and Porphysome accumulation in fluorescent VX2 (+) tissue compared to fluorescent VX2 (-) and non-fluorescent VX2 (-) tissue. Results: Of 17 VX2 models, 10 received 4 mg/kg of Porphysomes and 7 received 1 mg/kg. Seventeen tumours (UT), 81 lymph nodes (LN) and 54 abdominal metastases (AM) were fluorescence-positive and resected. Of these, 17 UT, 60 LN and 45 AM were VX2 (+), while 16 LN and 5 AM were VX2 (-). Nine specimens were excluded from analysis. Thirty-one LN and 53 peritoneal biopsies were fluorescence-negative and resected. Of these, all LN and 51/53 biopsies were VX2 (-) with only 2 false-negative biopsies. Sensitivity and specificity of PYRO-FGR for VX2 (+) tissue was 98.4% / 80.0% overall, 100% / 100% for UT, 100% / 66.0 % for LN and 95.7% / 91.4% for AM. Increased SBR and biodistribution was observed in VX2 (+) tissue vs. VX2 (-) tissue. Conclusions: Porphysomes are a highly sensitive imaging agent for intra-operative detection and resection of uterine tumour, metastatic lymph nodes and abdominal metastases.
Collapse
|
30
|
Shao S, Rajendiran V, Lovell JF. Metalloporphyrin Nanoparticles: Coordinating Diverse Theranostic Functions. Coord Chem Rev 2019; 379:99-120. [PMID: 30559508 PMCID: PMC6294123 DOI: 10.1016/j.ccr.2017.09.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Metalloporphyrins serve key roles in natural biological processes and also have demonstrated utility for biomedical applications. They can be encapsulated or grafted in conventional nanoparticles or can self-assemble themselves at the nanoscale. A wide range of metals can be stably chelated either before or after porphyrin nanoparticle formation, without the necessity of any additional chelator chemistry. The addition of metals can substantially alter a range of behaviors such as modulating phototherapeutic efficacy; conferring responsiveness to biological stimuli; or providing contrast for magnetic resonance, positron emission or surface enhanced Raman imaging. Chelated metals can also provide a convenient handle for bioconjugation with other molecules via axial coordination. This review provides an overview of some recent biomedical, nanoparticulate approaches involving gain-of-function metalloporphyrins and related molecules.
Collapse
Affiliation(s)
- Shuai Shao
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| | - Venugopal Rajendiran
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
- Department of Chemistry, School of Basic and Applied Sciences, Central University of Tamil Nadu, Thiruvarur 610 005, India
| | - Jonathan F. Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, USA
| |
Collapse
|
31
|
Massiot J, Rosilio V, Makky A. Photo-triggerable liposomal drug delivery systems: from simple porphyrin insertion in the lipid bilayer towards supramolecular assemblies of lipid–porphyrin conjugates. J Mater Chem B 2019; 7:1805-1823. [DOI: 10.1039/c9tb00015a] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Light-responsive liposomes are considered nowadays as one of the most promising nanoparticulate systems for the delivery and release of an active pharmaceutical ingredient (API) in a spatio-temporal manner.
Collapse
Affiliation(s)
- Julien Massiot
- Institut Galien Paris Sud
- Univ Paris-Sud
- CNRS
- Université Paris-Saclay
- 92296 Châtenay-Malabry
| | - Véronique Rosilio
- Institut Galien Paris Sud
- Univ Paris-Sud
- CNRS
- Université Paris-Saclay
- 92296 Châtenay-Malabry
| | - Ali Makky
- Institut Galien Paris Sud
- Univ Paris-Sud
- CNRS
- Université Paris-Saclay
- 92296 Châtenay-Malabry
| |
Collapse
|
32
|
|
33
|
Hajdu I, Makhlouf A, Solomon VR, Michel D, Al-Dulaymi M, Wasan KM, Fonge H, Badea I. A 89Zr-labeled lipoplex nanosystem for image-guided gene delivery: design, evaluation of stability and in vivo behavior. Int J Nanomedicine 2018; 13:7801-7818. [PMID: 30538460 PMCID: PMC6257135 DOI: 10.2147/ijn.s179806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background With the advances in radiopharmaceutical research, the development of image-guided therapy has become a major interest. While the development of theranostic nanotherapeutics is frequently associated with cancer chemotherapy, phototherapy and radiotherapy, there is little information available on the in vivo monitoring of gene delivery systems and the application of image-guided approach in gene therapy. The goal of this work was to determine the in vivo behavior of DNA delivery nanosystems - based on cationic gemini surfactants – designed for image-guided gene therapy. We tested the feasibility of monitoring tumor accumulation of gene delivery nanoparticles by positron emission tomography. Methods To be able to conjugate radiotracers to the nanoparticles, a deferoxamine-modified gemini surfactant was synthesized, DNA-containing lipoplex nanoparticles were formulated, and radiolabeled with Zirconium-89 (89Zr). The pharmacokinetics and biodistribution of 89Zr labeled surfactant and 89Zr labeled nanoparticles were monitored in mice by microPET/CT imaging and ex vivo gamma counting. Results Modification of the nanoparticles with deferoxamine did not alter their physicochemical properties. The radiolabeled nanoparticles (labeling efficiency of 95±3%) were stable in PBS and serum. The biological half-life of the 89Zr labeled nanoparticles was significantly higher compared to 89Zr labeled surfactant. As expected, the nanoparticles had significantly higher liver accumulation than the radiolabeled surfactant alone and lower kidney accumulation. Tumor uptake was detected at 2 hours post injection and decreased throughout the 3-day monitoring. Conclusion We propose that radiolabeling DNA delivery lipoplex nanosystems is a promising approach for the design and optimization of image-guided nanomedicines, especially in the context of cancer gene therapy.
Collapse
Affiliation(s)
- Istvan Hajdu
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Amal Makhlouf
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada, .,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, 12411 Cairo, Egypt
| | - Viswas Raja Solomon
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada,
| | - Deborah Michel
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Mays Al-Dulaymi
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Kishor M Wasan
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| | - Humphrey Fonge
- Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada, .,Department of Medical Imaging, Royal University Hospital Saskatoon, SK S7N 0W8, Canada,
| | - Ildiko Badea
- Drug Discovery and Development Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada,
| |
Collapse
|
34
|
Gao X, Guo L, Li J, Thu HE, Hussain Z. Nanomedicines guided nanoimaging probes and nanotherapeutics for early detection of lung cancer and abolishing pulmonary metastasis: Critical appraisal of newer developments and challenges to clinical transition. J Control Release 2018; 292:29-57. [PMID: 30359665 DOI: 10.1016/j.jconrel.2018.10.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 01/13/2023]
Abstract
Lung cancer (LC) is the second most prevalent type of cancer and primary cause of mortality among both men and women, worldwide. The most commonly employed diagnostic modalities for LC include chest X-ray (CXR), magnetic-resonance-imaging (MRI), computed tomography (CT-scan), and fused-positron-emitting-tomography-CT (PET-CT). Owing to several limitations associated with the use of conventional diagnostic tools such as radiation burden to the patient, misleading diagnosis ("missed lung cancer"), false staging and low sensitivity and resolution, contemporary diagnostic regimen needed to be employed for screening of LC. In recent decades, nanotechnology-guided interventions have been transpired as emerging nanoimaging probes for detection of LC at advanced stages, while producing signal amplification, better resolution for surface and deep tissue imaging, and enhanced translocation and biodistribution of imaging probes within the cancerous tissues. Besides enormous potential of nanoimaging probes, nanotechnology-based advancements have also been evidenced for superior efficacy for treatment of LC and abolishing pulmonary metastasis (PM). The success of nanotherapeutics is due to their ability to maximise translocation and biodistribution of anti-neoplastic agents into the tumor tissues, improve pharmacokinetic profiles of anti-metastatic agents, optimise target-specific drug delivery, and control release kinetics of encapsulated moieties in target tissues. This review aims to overview and critically discuss the superiority of nanoimaging probes and nanotherapeutics over conventional regimen for early detection of LC and abolishing PM. Current challenges to clinical transition of nanoimaging probes and therapeutic viability of nanotherapeutics for treatment for LC and PM have also been pondered.
Collapse
Affiliation(s)
- Xiaoling Gao
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Lihua Guo
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jianqiang Li
- Department of Respiratory and Critical Care Medicine, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, China
| | - Hnin Ei Thu
- Department of Pharmacology and Dental Therapeutics, Faculty of Dentistry, Lincoln University College, Jalan Stadium, SS 7/15, Kelana Jaya, 47301 Petaling Jaya, Selangor, Malaysia
| | - Zahid Hussain
- Department of Pharmaceutics, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM) Selangor, Puncak Alam Campus, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| |
Collapse
|
35
|
Ujiie H, Ding L, Fan R, Kato T, Lee D, Fujino K, Kinoshita T, Lee CY, Waddell TK, Keshavjee S, Wilson BC, Zheng G, Chen J, Yasufuku K. Porphyrin-High-Density Lipoprotein: A Novel Photosensitizing Nanoparticle for Lung Cancer Therapy. Ann Thorac Surg 2018; 107:369-377. [PMID: 30316853 DOI: 10.1016/j.athoracsur.2018.08.053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/20/2018] [Accepted: 08/22/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND We have developed ultrasmall porphyrin-high-density lipoprotein (HDL) nanoparticles (<20 nm), called "porphyrinHDL," that have a high density of porphyrin molecules and dissociate rapidly upon tumor cell accumulation to become fluorescent and photoactive. This is introduced as a novel activatable photosensitizer for image-guided photodynamic therapy (PDT). Here, we report the studies of these nanoparticles targeted to scavenger receptor class B type I (SR-BI) expressed on lung cancer cells as a first step toward development of a minimally invasive treatment for peripheral lung cancer and metastatic lymph nodes of advanced lung cancer. METHODS The in vitro uptake of porphyrinHDL and the corresponding PDT efficacy were evaluated in both SR-BI-positive and SR-BI-negative lung cancer cell lines. A clinically relevant orthotopic lung cancer model in mice was used to examine fluorescence activation and quantification of uptake in tumor. In addition, we investigated the effect of porphyrinHDL-mediated PDT. RESULTS PorphyrinHDL promoted proper intracellular uptake in the H460 human lung cancer cell line. When irradiated with a 671-nm PDT laser, porphyrinHDL produced significant therapeutic effectiveness in vitro. After systemic administration in mice with orthotopic lung cancer xenografts, porphyrinHDL demonstrated selective accumulation and photoactivation in tumor with significantly enhanced disease-to-normal tissue contrast. Moreover, porphyrinHDL-PDT significantly induced cell apoptosis in lung tumors (73.2%) without toxicity in normal tissues or damage to adjacent critical structures. CONCLUSIONS SR-BI-targeted porphyrinHDL-mediated PDT of lung cancer is selective and effective in vitro and in vivo. These initial proof-of-principle studies suggest the potential of a "smart" PDT approach for highly selective tumor ablation.
Collapse
Affiliation(s)
- Hideki Ujiie
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Lili Ding
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Rong Fan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Tatsuya Kato
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Daiyoon Lee
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Kosuke Fujino
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Tomonari Kinoshita
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Chang Young Lee
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Thomas K Waddell
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Shaf Keshavjee
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario
| | - Brian C Wilson
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario; Department of Medical Biophysics, University of Toronto, Toronto, Ontario
| | - Gang Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario; Department of Medical Biophysics, University of Toronto, Toronto, Ontario; Guided Therapeutics, Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario; Institute of Biomaterial and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Juan Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Kazuhiro Yasufuku
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario; Department of Medical Biophysics, University of Toronto, Toronto, Ontario; Guided Therapeutics, Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario; Institute of Biomaterial and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Kleynhans J, Grobler AF, Ebenhan T, Sathekge MM, Zeevaart JR. Radiopharmaceutical enhancement by drug delivery systems: A review. J Control Release 2018; 287:177-193. [DOI: 10.1016/j.jconrel.2018.08.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/02/2018] [Accepted: 08/03/2018] [Indexed: 12/17/2022]
|
37
|
Sandland J, Malatesti N, Boyle R. Porphyrins and related macrocycles: Combining photosensitization with radio- or optical-imaging for next generation theranostic agents. Photodiagnosis Photodyn Ther 2018; 23:281-294. [DOI: 10.1016/j.pdpdt.2018.06.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/22/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
|
38
|
Tang WL, Tang WH, Li SD. Cancer theranostic applications of lipid-based nanoparticles. Drug Discov Today 2018; 23:1159-1166. [DOI: 10.1016/j.drudis.2018.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/18/2022]
|
39
|
Abstract
A recent metaanalysis shows that 0.7% of nanoparticles are delivered to solid tumors. This low delivery efficiency has major implications in the translation of cancer nanomedicines, as most of the nanomedicines are sequestered by nontumor cells. To improve the delivery efficiency, there is a need to investigate the quantitative contribution of each organ in blocking the transport of nanoparticles to solid tumors. Here, we hypothesize that the removal of the liver macrophages, cells that have been reported to take up the largest amount of circulating nanoparticles, would lead to a significant increase in the nanoparticle delivery efficiency to solid tumors. We were surprised to discover that the maximum achievable delivery efficiency was only 2%. In our analysis, there was a clear correlation between particle design, chemical composition, macrophage depletion, tumor pathophysiology, and tumor delivery efficiency. In many cases, we observed an 18-150 times greater delivery efficiency, but we were not able to achieve a delivery efficiency higher than 2%. The results suggest the need to look deeper at other organs such as the spleen, lymph nodes, and tumor in mediating the delivery process. Systematically mapping the contribution of each organ quantitatively will allow us to pinpoint the cause of the low tumor delivery efficiency. This, in effect, enables the generation of a rational strategy to improve the delivery efficiency of nanoparticles to solid tumors either through the engineering of multifunctional nanosystems or through manipulation of biological barriers.
Collapse
|
40
|
Molecular imaging in drug development: Update and challenges for radiolabeled antibodies and nanotechnology. Methods 2017; 130:23-35. [DOI: 10.1016/j.ymeth.2017.07.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/08/2017] [Accepted: 07/18/2017] [Indexed: 01/01/2023] Open
|
41
|
Gao W, Li S, Liu Z, Sun Y, Cao W, Tong L, Cui G, Tang B. Targeting and destroying tumor vasculature with a near-infrared laser-activated "nanobomb" for efficient tumor ablation. Biomaterials 2017; 139:1-11. [PMID: 28578297 DOI: 10.1016/j.biomaterials.2017.05.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/24/2017] [Accepted: 05/21/2017] [Indexed: 12/11/2022]
Abstract
Attacking the supportive vasculature network of a tumor offers an important new avenue for cancer therapy. Herein, a near-infrared (NIR) laser-activated "nanobomb" was developed as a noninvasive and targeted physical therapeutic strategy to effectively disrupt tumor neovasculature in an accurate and expeditious manner. This "nanobomb" was rationally fabricated via the encapsulation of vinyl azide (VA) into c(RGDfE) peptide-functionalized, hollow copper sulfide (HCuS) nanoparticles. The resulting RGD@HCuS(VA) was selectively internalized into integrin αvβ3-expressing tumor vasculature endothelial cells and dramatically increased the photoacoustic signals from the tumor neovasculature, achieving a maximum signal-to-noise ratio at 4 h post-injection. Upon NIR irradiation, the local temperature increase triggered VA to release N2 bubbles rapidly. Subsequently, these N2 bubbles could instantly explode to destroy the neovasculature and further induce necrosis of the surrounding tumor cells. A single-dose injection of RGD@HCuS(VA) led to complete tumor regression after laser irradiation, with no tumor regrowth for 30 days. More importantly, high-resolution photoacoustic angiography, combined with excellent biodegradability, facilitated the precise destruction of tumor neovasculature by RGD@HCuS(VA) without damaging normal tissues. These results demonstrate the great potential of this "nanobomb" for clinical translation to treat cancer patients with NIR laser-accessible orthotopic tumors.
Collapse
Affiliation(s)
- Wen Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Shuangshuang Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Zhenhua Liu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Yuhui Sun
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Wenhua Cao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Lili Tong
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Guanwei Cui
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan 250014, PR China.
| |
Collapse
|
42
|
Rajora MA, Ding L, Valic M, Jiang W, Overchuk M, Chen J, Zheng G. Tailored theranostic apolipoprotein E3 porphyrin-lipid nanoparticles target glioblastoma. Chem Sci 2017; 8:5371-5384. [PMID: 28970916 PMCID: PMC5609152 DOI: 10.1039/c7sc00732a] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/18/2017] [Indexed: 12/21/2022] Open
Abstract
Size-controlled discoidal and cholesteryl oleated-loaded spherical, intrinsically multimodal porphyrin-lipid nanoparticles targeted glioblastoma via apoE3 and LDLR.
The development of curative glioblastoma treatments and tumour-specific contrast agents that can overcome the blood–brain barrier (BBB) and infiltrative tumour morphology remains a challenge. Apolipoprotein E3 (apoE3) is a high density lipoprotein apolipoprotein that chaperones the transcytosis of nanoparticles across the BBB, and displays high-affinity binding with the low density lipoprotein receptor (LDLR), a cell-surface receptor overexpressed by glioblastoma cells. This LDLR overexpression and apoE3 binding capacity was exploited for the development of glioblastoma-targeted porphyrin-lipid apoE3 lipid nanoparticles (pyE-LNs) with intrinsic theranostic properties. Size-controlled discoidal and cholesteryl oleate (CO)-loaded spherical pyE-LNs were synthesized through the systematic variation of particle composition, which dictated nanoparticle size and morphology. Composition optimization yielded 30 nm pyE-LNs with stable loading of apoE3 and porphyrin-lipid that simultaneously conferred the nanoparticles with glioblastoma targeting and activatable near-infrared fluorescence imaging functionalities. A 4-fold higher uptake of pyE-LNs by LDLR-expressing U87 glioblastomas cells relative to minimally expressing ldlA7 cells was observed in vitro. This uptake was a result of receptor-mediated endocytosis, which could be inhibited through LDL competition and acetylation of particle apoE3 moieties. ApoE3-dependent delivery of pyE-LN to glioblastomas was also demonstrated in orthotopic U87-GFP tumour-bearing animals. Quantification of CO-loaded pyE-LN biodistribution demonstrated successful selective uptake of porphyrin by malignant tissue, with a 4 : 1 tumour : healthy tissue particle specificity. This allowed for the detection of strong, tumour-localized porphyrin fluorescence, which was diminished when apoE3-devoid py-LN particles were administered. Furthermore, this selective uptake yielded cell-specific potent PDT sensitization in vitro, resulting in an 83% reduction in glioblastoma cell viability. These results highlight the promising capacity of pyE-LNs to target porphyrin delivery to glioblastoma tumours for theranostic applications.
Collapse
Affiliation(s)
- M A Rajora
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada . .,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - L Ding
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada .
| | - M Valic
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada . .,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - W Jiang
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada .
| | - M Overchuk
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada . .,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada
| | - J Chen
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada .
| | - G Zheng
- Princess Margaret Cancer Centre , University Health Network , 101 College Street , Toronto , Ontario M5G 1L7 , Canada . .,Institute of Biomaterials and Biomedical Engineering , University of Toronto , 164 College Street , Toronto , Ontario M5S 3G9 , Canada.,Department of Medical Biophysics , University of Toronto , 101 College Street , Toronto , Ontario M5G 1L7 , Canada
| |
Collapse
|
43
|
Goel S, England CG, Chen F, Cai W. Positron emission tomography and nanotechnology: A dynamic duo for cancer theranostics. Adv Drug Deliv Rev 2017; 113:157-176. [PMID: 27521055 PMCID: PMC5299094 DOI: 10.1016/j.addr.2016.08.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/29/2016] [Accepted: 08/03/2016] [Indexed: 12/18/2022]
Abstract
Development of novel imaging probes for cancer diagnosis is critical for early disease detection and management. The past two decades have witnessed a surge in the development and evolution of radiolabeled nanoparticles as a new frontier in personalized cancer nanomedicine. The dynamic synergism of positron emission tomography (PET) and nanotechnology combines the sensitivity and quantitative nature of PET with the multifunctionality and tunability of nanomaterials, which can help overcome certain key challenges in the field. In this review, we discuss the recent advances in radionanomedicine, exemplifying the ability to tailor the physicochemical properties of nanomaterials to achieve optimal in vivo pharmacokinetics and targeted molecular imaging in living subjects. Innovations in development of facile and robust radiolabeling strategies and biomedical applications of such radionanoprobes in cancer theranostics are highlighted. Imminent issues in clinical translation of radiolabeled nanomaterials are also discussed, with emphasis on multidisciplinary efforts needed to quickly move these promising agents from bench to bedside.
Collapse
Affiliation(s)
- Shreya Goel
- Materials Science Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Christopher G England
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Feng Chen
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA.
| | - Weibo Cai
- Materials Science Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Radiology, University of Wisconsin-Madison, Madison, WI 53792, USA; University of Wisconsin Carbone Cancer Center, Madison, WI 53792, USA.
| |
Collapse
|
44
|
Huang H, Lovell JF. Advanced Functional Nanomaterials for Theranostics. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1603524. [PMID: 28824357 PMCID: PMC5560626 DOI: 10.1002/adfm.201603524] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Nanoscale materials have been explored extensively as agents for therapeutic and diagnostic (i.e. theranostic) applications. Research efforts have shifted from exploring new materials in vitro to designing materials that function in more relevant animal disease models, thereby increasing potential for clinical translation. Current interests include non-invasive imaging of diseases, biomarkers and targeted delivery of therapeutic drugs. Here, we discuss some general design considerations of advanced theranostic materials and challenges of their use, from both diagnostic and therapeutic perspectives. Common classes of nanoscale biomaterials, including magnetic nanoparticles, quantum dots, upconversion nanoparticles, mesoporous silica nanoparticles, carbon-based nanoparticles and organic dye-based nanoparticles, have demonstrated potential for both diagnosis and therapy. Variations such as size control and surface modifications can modulate biocompatibility and interactions with target tissues. The needs for improved disease detection and enhanced chemotherapeutic treatments, together with realistic considerations for clinically translatable nanomaterials will be key driving factors for theranostic agent research in the near future.
Collapse
Affiliation(s)
- Haoyuan Huang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, United States
| |
Collapse
|
45
|
Zhao J, Zhou M, Li C. Synthetic nanoparticles for delivery of radioisotopes and radiosensitizers in cancer therapy. Cancer Nanotechnol 2016; 7:9. [PMID: 27909463 PMCID: PMC5112292 DOI: 10.1186/s12645-016-0022-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Radiotherapy has been, and will continue to be, a critical modality to treat cancer. Since the discovery of radiation-induced cytotoxicity in the late 19th century, both external and internal radiation sources have provided tremendous benefits to extend the life of cancer patients. Despite the dramatic improvement of radiation techniques, however, one challenge persists to limit the anti-tumor efficacy of radiotherapy, which is to maximize the deposited dose in tumor while sparing the rest of the healthy vital organs. Nanomedicine has stepped into the spotlight of cancer diagnosis and therapy during the past decades. Nanoparticles can potentiate radiotherapy by specifically delivering radionuclides or radiosensitizers into tumors, therefore enhancing the efficacy while alleviating the toxicity of radiotherapy. This paper reviews recent advances in synthetic nanoparticles for radiotherapy and radiosensitization, with a focus on the enhancement of in vivo anti-tumor activities. We also provide a brief discussion on radiation-associated toxicities as this is an area that, up to date, has been largely missing in the literature and should be closely examined in future studies involving nanoparticle-mediated radiosensitization.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Road, Houston, TX 77054 USA
| | - Min Zhou
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009 Zhejiang China
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, 1881 East Road, Houston, TX 77054 USA
| |
Collapse
|
46
|
Lee SB, Yoon G, Lee SW, Jeong SY, Ahn BC, Lim DK, Lee J, Jeon YH. Combined Positron Emission Tomography and Cerenkov Luminescence Imaging of Sentinel Lymph Nodes Using PEGylated Radionuclide-Embedded Gold Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:4894-4901. [PMID: 27439987 DOI: 10.1002/smll.201601721] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Indexed: 06/06/2023]
Abstract
New imaging probes with high sensitivity and stability are urgently needed to accurately detect sentinel lymph nodes (SLNs) for successful cancer diagnosis. Herein, the use of highly sensitive and stable PEGylated radionuclide-embedded gold nanoparticles (PEG-RIe-AuNPs) is reported for the detection of SLNs by combined positron emission tomography and Cerenkov luminescence imaging (PET/CLI). PEG-RIe-AuNPs show high sensitivity and stability both in vitro and in vivo, and are not toxic to normal ovarian and immune cells. In vivo PET/CLI imaging clearly reveals SLNs as early as 1 h post PEG-RIe-AuNP-injection, with peak signals achieved at 6 h postinjection, which is consistent with the biodistribution results. Taken together, the data provide strong evidence that PEG-RIe-AuNPs are promising as potential lymphatic tracers in biomedical imaging for pre and intraoperative surgical guidance.
Collapse
Affiliation(s)
- Sang Bong Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 702-210, South Korea
| | - GhilSuk Yoon
- Department of Pathology, Kyungpook National University Hospital, Daegu, 702-210, South Korea
| | - Sang-Woo Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 702-210, South Korea
| | - Shin Young Jeong
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea.
| | - Jaetae Lee
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea.
- Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, 360-4, South Korea.
| | - Yong Hyun Jeon
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, 702-210, South Korea.
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, 702-210, South Korea.
| |
Collapse
|
47
|
Jin CS, Overchuk M, Cui L, Wilson BC, Bristow RG, Chen J, Zheng G. Nanoparticle-Enabled Selective Destruction of Prostate Tumor Using MRI-Guided Focal Photothermal Therapy. Prostate 2016; 76:1169-81. [PMID: 27198587 DOI: 10.1002/pros.23203] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/27/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The Magnetic Resonance Imaging (MRI)-guided focal laser therapy has shown early promise in Phase 1 trial treating low/intermediate-risk localized prostate cancer (PCa), but the lack of tumor selectivity and low efficiency of heat generation remain as drawbacks of agent-free laser therapy. Intrinsic multifunctional porphyrin-nanoparticles (porphysomes) have been exploited to treat localized PCa by MRI-guided focal photothermal therapy (PTT) with significantly improved efficiency and tumor selectivity over prior methods of PTT, providing an effective and safe alternative to active surveillance or radical therapy. METHODS The tumor accumulation of porphysomes chelated with copper-64 was determined and compared with the clinic standard (18) F-FDG in an orthotropic PCa mouse model by positron emission tomography (PET) imaging, providing quantitative assessment for PTT dosimetry. The PTT was conducted with MRI-guided light delivery and monitored by MR thermometry, mimicking the clinical protocol. The efficacy of treatment and adverse effects to surround tissues were evaluated by histology analysis and tumor growth in survival study via MRI. RESULTS Porphysomes showed superior tumor-to-prostate selectivity over (18) F-FDG (6:1 vs. 0.36:1). MR thermometry detected tumor temperature increased to ≥55°C within 2 min (671 nm at 500 mW), but minimal increase in surrounding tissues. Porphysome enabled effective PTT eradication of tumor without damaging adjacent organs in orthotropic PCa mouse model. CONCLUSIONS Porphysome-enabled MRI-guided focal PTT could be an effective and safe approach to treat PCa at low risk of progression, thus addressing the significant unmet clinical needs and benefiting an ever-growing number of patients who may be over-treated and risk unnecessary side effects from radical therapies. Prostate 76:1169-1181, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Cheng S Jin
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Marta Overchuk
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Liyang Cui
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Medical Isotopes Research Center, Peking University, Beijing, China
| | - Brian C Wilson
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Robert G Bristow
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Juan Chen
- Princess Margaret Cancer Center, UHN, Toronto, Canada
| | - Gang Zheng
- Princess Margaret Cancer Center, UHN, Toronto, Canada
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
48
|
Jin CS, Wada H, Anayama T, McVeigh PZ, Hu HP, Hirohashi K, Nakajima T, Kato T, Keshavjee S, Hwang D, Wilson BC, Zheng G, Yasufuku K. An Integrated Nanotechnology-Enabled Transbronchial Image-Guided Intervention Strategy for Peripheral Lung Cancer. Cancer Res 2016; 76:5870-5880. [PMID: 27543602 DOI: 10.1158/0008-5472.can-15-3196] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 06/07/2016] [Indexed: 12/25/2022]
Abstract
Early detection and efficient treatment modality of early-stage peripheral lung cancer is essential. Current nonsurgical treatments for peripheral lung cancer show critical limitations associated with various complications, requiring alternative minimally invasive therapeutics. Porphysome nanoparticle-enabled fluorescence-guided transbronchial photothermal therapy (PTT) of peripheral lung cancer was developed and demonstrated in preclinical animal models. Systemically administered porphysomes accumulated in lung tumors with significantly enhanced disease-to-normal tissue contrast, as confirmed in three subtypes of orthotopic human lung cancer xenografts (A549, H460, and H520) in mice and in an orthotopic VX2 tumor in rabbits. An in-house prototype fluorescence bronchoscope demonstrated the capability of porphysomes for in vivo imaging of lung tumors in the mucosal/submucosal layers, providing real-time fluorescence guidance for transbronchial PTT. Porphysomes also enhanced the efficacy of transbronchial PTT significantly and resulted in selective and efficient tumor tissue ablation in the rabbit model. A clinically used cylindrical diffuser fiber successfully achieved tumor-specific thermal ablation, showing promising evidence for the clinical translation of this novel platform to impact upon nonsurgical treatment of early-stage peripheral lung cancer. Cancer Res; 76(19); 5870-80. ©2016 AACR.
Collapse
Affiliation(s)
- Cheng S Jin
- Graduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. Institute of Biomaterial and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario, Canada
| | - Hironobu Wada
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Takashi Anayama
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Patrick Z McVeigh
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Hsin Pei Hu
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Kentaro Hirohashi
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Takahiro Nakajima
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Tatsuya Kato
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - David Hwang
- Division of Experimental Therapeutics, Respiratory & Critical Care, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Brian C Wilson
- Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gang Zheng
- Graduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada. Institute of Biomaterial and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada. Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario, Canada. Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Kazuhiro Yasufuku
- Princess Margaret Cancer Centre and TECHNA Institute, University Health Network, Toronto, Ontario, Canada. Division of Thoracic Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
MacLaughlin CM, Ding L, Jin C, Cao P, Siddiqui I, Hwang DM, Chen J, Wilson BC, Zheng G, Hedley DW. Porphysome nanoparticles for enhanced photothermal therapy in a patient-derived orthotopic pancreas xenograft cancer model: a pilot study. JOURNAL OF BIOMEDICAL OPTICS 2016; 21:84002. [PMID: 27552306 DOI: 10.1117/1.jbo.21.8.084002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 08/02/2016] [Indexed: 05/15/2023]
Abstract
Local disease control is a major challenge in pancreatic cancer treatment, because surgical resection of the primary tumor is only possible in a minority of patients and radiotherapy cannot be delivered in curative doses. Despite the promise of photothermal therapy (PTT) for focal ablation of pancreatic tumors, this approach remains underinvestigated. Using photothermal sensitizers in combination with laser light irradiation for PTT can result in more efficient conversion of light energy to heat and improved spatial confinement of thermal destruction to the tumor. Porphysomes are self-assembled nanoparticles composed mainly of pyropheophorbide-conjugated phospholipids, enabling the packing of ∼80,000 porphyrin photosensitizers per particle. The high-density porphyrin loading imparts enhanced photonic properties and enables high-payload tumor delivery. A patient-derived orthotopic pancreas xenograft model was used to evaluate the feasibility of porphysome-enhanced PTT for pancreatic cancer. Biodistribution and tumor accumulation were evaluated using fluorescence intensity measurements from homogenized tissues and imaging of excised organs. Tumor surface temperature was recorded using IR optical imaging during light irradiation to monitor treatment progress. Histological analyses were conducted to determine the extent of PTT thermal damage. These studies may provide insight into the influence of heat-sink effect on thermal therapy dosimetry for well-perfused pancreatic tumors.
Collapse
Affiliation(s)
- Christina M MacLaughlin
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, CanadabUniversity of Toronto, Department of Medical Biophysics, 101 College Street, Toronto, Ontario M5G 1L7, CanadacPrincess Margaret Hospital, Department of Medical Oncology and Hematology, 610 University Avenue, Toronto, Ontario M5T 2M9, Canada
| | - Lili Ding
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Cheng Jin
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, CanadabUniversity of Toronto, Department of Medical Biophysics, 101 College Street, Toronto, Ontario M5G 1L7, CanadadUniversity of Toronto, Department of Pharmaceutical Sciences, 144 College Street, Toronto, Ontario M5T 2M9, Canada
| | - Pingjiang Cao
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Iram Siddiqui
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - David M Hwang
- University Health Network, Department of Pathology, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Juan Chen
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Brian C Wilson
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, CanadabUniversity of Toronto, Department of Medical Biophysics, 101 College Street, Toronto, Ontario M5G 1L7, CanadacPrincess Margaret Hospital, Department of Medical Oncology and Hematology, 610 University Avenue, Toronto, Ontario M5T 2M9, Canada
| | - Gang Zheng
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, CanadabUniversity of Toronto, Department of Medical Biophysics, 101 College Street, Toronto, Ontario M5G 1L7, CanadadUniversity of Toronto, Department of Pharmaceutical Sciences, 144 College Street, Toronto, Ontario M5T 2M9, Canada
| | - David W Hedley
- University Health Network, Princess Margaret Cancer Center, 101 College Street, Toronto, Ontario M5G 1L7, CanadabUniversity of Toronto, Department of Medical Biophysics, 101 College Street, Toronto, Ontario M5G 1L7, CanadacPrincess Margaret Hospital, Department of Medical Oncology and Hematology, 610 University Avenue, Toronto, Ontario M5T 2M9, Canada
| |
Collapse
|
50
|
Zhou Y, Liang X, Dai Z. Porphyrin-loaded nanoparticles for cancer theranostics. NANOSCALE 2016; 8:12394-12405. [PMID: 26730838 DOI: 10.1039/c5nr07849k] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Porphyrins have been used as pioneering theranostic agents not only for the photodynamic therapy, sonodynamic therapy and radiotherapy of cancer, but also for diagnostic fluorescence imaging, magnetic resonance imaging and photoacoustic imaging. A variety of porphyrins have been developed but very few of them have actually been employed in clinical trials due to their poor selectivity to tumorous tissue and high accumulation rates in the skin. In addition, most porphyrin molecules are hydrophobic and form aggregates in aqueous media. Nevertheless, the use of nanoparticles as porphyrin carriers shows great promise to overcome these shortcomings. Encapsulating or attaching porphyrins to nanoparticles makes them more suitable for tissue delivery because we can create materials with a conveniently specific tissue lifetime, specific targeting, immune tolerance, and hydrophilicity as well as other characteristics through rational design. In addition, various functional components (e.g. for targeting, imaging or therapeutic functions) can be easily introduced into a single nanoparticle platform for cancer theranostics. This review presents the current state of knowledge on porphyrin-loaded nanoparticles for the interwined imaging and therapy of cancer. The future trends and limitations of prophyrin-loaded nanoparticles are also outlined.
Collapse
Affiliation(s)
- Yiming Zhou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China.
| | | | | |
Collapse
|