1
|
Kuznetsov KM, Cariou K, Gasser G. Two in one: merging photoactivated chemotherapy and photodynamic therapy to fight cancer. Chem Sci 2024:d4sc04608k. [PMID: 39464604 PMCID: PMC11499979 DOI: 10.1039/d4sc04608k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
The growing number of cancer cases requires the development of new approaches for treatment. A therapy that has attracted the special attention of scientists is photodynamic therapy (PDT) due to its spatial and temporal resolution. However, it is accepted that this treatment methodology has limited application in cases of low cellular oxygenation, which is typical of cancerous tissues. Therefore, a strategy to overcome this drawback has been to combine this therapy with photoactivated chemotherapy (PACT), which works independently of the presence of oxygen. In this perspective, we examine compounds that act as both PDT and PACT agents and summarize their photophysical and biological characteristics.
Collapse
Affiliation(s)
- Kirill M Kuznetsov
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| | - Kevin Cariou
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology 75005 Paris France http://www.gassergroup.com/ +33 1 85 78 41 51
| |
Collapse
|
2
|
Giram P, Bist G, Woo S, Wohlfert E, Pili R, You Y. Prodrugs of paclitaxel improve in situ photo-vaccination. Photochem Photobiol 2024:10.1111/php.14025. [PMID: 39384406 PMCID: PMC11978925 DOI: 10.1111/php.14025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/22/2024] [Indexed: 10/11/2024]
Abstract
Photodynamic therapy (PDT) effectively kills cancer cells and initiates immune responses that promote anticancer effects locally and systemically. Primarily developed for local and regional cancers, the potential of PDT for systemic antitumor effects [in situ photo-vaccination (ISPV)] remains underexplored. This study investigates: (1) the comparative effectiveness of paclitaxel (PTX) prodrug [Pc-(L-PTX)2] for PDT and site-specific PTX effects versus its pseudo-prodrug [Pc-(NCL-PTX)2] for PDT combined with checkpoint inhibitors; (2) mechanisms driving systemic antitumor effects; and (3) the prophylactic impact on preventing cancer recurrence. A bilateral tumor model was established in BALB/c mice through subcutaneous injection of CT26 cells. Mice received the PTX prodrug (0.5 μmole kg-1, i.v.), and tumors were treated with a 690-nm laser (75 mW cm-2 for 30 min, drug-light interval 0.5 h, light does 135 J cm-1), followed by anti-CTLA-4 (100 μg dose-1, i.p.) on days 1, 4, and 7. Notable enhancement in both local and systemic antitumor effectiveness was observed with [Pc-(L-PTX)2] compared to [Pc-(NCL-PTX)2] with checkpoint inhibitor. Immune cell depletion and immunohistochemistry confirmed neutrophils and CD8+ T cells are effectors for systemic antitumor effects. Treatment-induced immune memory resisted newly rechallenged CT26, showcasing prophylactic benefits. ISPV with a PTX prodrug and anti-CTLA-4 is a promising approach for treating metastatic cancers and preventing recurrence.
Collapse
Affiliation(s)
- Prabhanjan Giram
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Ganesh Bist
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Elizabeth Wohlfert
- Department of Microbiology and Immunology, University at Buffalo School of Medicine, Buffalo, NY,14203, USA
| | - Roberto Pili
- Division of Hematology and Oncology, Department of Medicine, University at Buffalo, Buffalo, NY, 14203, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| |
Collapse
|
3
|
Kita K, Burdowski A. Recent clinical trials and optical control as a potential strategy to develop microtubule-targeting drugs in colorectal cancer management. World J Gastroenterol 2024; 30:1780-1790. [PMID: 38659489 PMCID: PMC11036503 DOI: 10.3748/wjg.v30.i13.1780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/08/2024] [Accepted: 03/19/2024] [Indexed: 04/03/2024] Open
Abstract
Colorectal cancer (CRC) has remained the second and the third leading cause of cancer-related death worldwide and in the United States, respectively. Although significant improvement in overall survival has been achieved, death in adult populations under the age of 55 appears to have increased in the past decades. Although new classes of therapeutic strategies such as immunotherapy have emerged, their application is very limited in CRC so far. Microtubule (MT) inhibitors such as taxanes, are not generally successful in CRC. There may be some way to make MT inhibitors work effectively in CRC. One potential advantage that we can take to treat CRC may be the combination of optical techniques coupled to an endoscope or other fiber optics-based devices. A combination of optical devices and photo-activatable drugs may allow us to locally target advanced CRC cells with highly potent MT-targeting drugs. In this Editorial review, we would like to discuss the potential of optogenetic approaches in CRC management.
Collapse
Affiliation(s)
- Katsuhiro Kita
- Department of Biology, St. Francis College, Brooklyn, NY 11201, United States
| | - Allen Burdowski
- Department of Biology, St. Francis College, Brooklyn, NY 11201, United States
| |
Collapse
|
4
|
Kuzmina NS, Fedotova EA, Jankovic P, Gribova GP, Nyuchev AV, Fedorov AY, Otvagin VF. Enhancing Precision in Photodynamic Therapy: Innovations in Light-Driven and Bioorthogonal Activation. Pharmaceutics 2024; 16:479. [PMID: 38675140 PMCID: PMC11053670 DOI: 10.3390/pharmaceutics16040479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/29/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Over the past few decades, photodynamic therapy (PDT) has evolved as a minimally invasive treatment modality offering precise control over cancer and various other diseases. To address inherent challenges associated with PDT, researchers have been exploring two promising avenues: the development of intelligent photosensitizers activated through light-induced energy transfers, charges, or electron transfers, and the disruption of photosensitive bonds. Moreover, there is a growing emphasis on the bioorthogonal delivery or activation of photosensitizers within tumors, enabling targeted deployment and activation of these intelligent photosensitive systems in specific tissues, thus achieving highly precise PDT. This concise review highlights advancements made over the last decade in the realm of light-activated or bioorthogonal photosensitizers, comparing their efficacy and shaping future directions in the advancement of photodynamic therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Alexey Yu. Fedorov
- Department of Organic Chemistry, Lobachevsky State University of Nizhny Novgorod, Gagarina Av. 23, 603950 Nizhny Novgorod, Russia; (N.S.K.); (E.A.F.); (P.J.); (G.P.G.); (A.V.N.)
| | - Vasilii F. Otvagin
- Department of Organic Chemistry, Lobachevsky State University of Nizhny Novgorod, Gagarina Av. 23, 603950 Nizhny Novgorod, Russia; (N.S.K.); (E.A.F.); (P.J.); (G.P.G.); (A.V.N.)
| |
Collapse
|
5
|
Chi T, Sang T, Wang Y, Ye Z. Cleavage and Noncleavage Chemistry in Reactive Oxygen Species (ROS)-Responsive Materials for Smart Drug Delivery. Bioconjug Chem 2024; 35:1-21. [PMID: 38118277 DOI: 10.1021/acs.bioconjchem.3c00476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The design and development of advanced drug delivery systems targeting reactive oxygen species (ROS) have gained significant interest in recent years for treating various diseases, including cancer, psychiatric diseases, cardiovascular diseases, neurological diseases, metabolic diseases, and chronic inflammations. Integrating specific chemical bonds capable of effectively responding to ROS and triggering drug release into the delivery system is crucial. In this Review, we discuss commonly used conjugation linkers (chemical bonds) and categorize them into two groups: cleavable linkers and noncleavable linkers. Our goal is to clarify their unique drug release mechanisms from a chemical perspective and provide practical organic synthesis approaches for their efficient production. We showcase numerous significant examples to demonstrate their synthesis routes and diverse applications. Ultimately, we strive to present a comprehensive overview of cleavage and noncleavage chemistry, offering insights into the development of smart drug delivery systems that respond to ROS.
Collapse
Affiliation(s)
- Teng Chi
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ting Sang
- School of Stomatology of Nanchang University & Jiangxi Province Clinical Research Center for Oral Diseases & The Key Laboratory of Oral Biomedicine, Nanchang 330006, China
| | - Yanjing Wang
- Department of Chemistry, Washington University, St. Louis, Missouri 63130, United States
| | - Zhou Ye
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong S.A.R. 999077, China
| |
Collapse
|
6
|
Fu Q, Shen S, Sun P, Gu Z, Bai Y, Wang X, Liu Z. Bioorthogonal chemistry for prodrug activation in vivo. Chem Soc Rev 2023; 52:7737-7772. [PMID: 37905601 DOI: 10.1039/d2cs00889k] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Prodrugs have emerged as a major strategy for addressing clinical challenges by improving drug pharmacokinetics, reducing toxicity, and enhancing treatment efficacy. The emergence of new bioorthogonal chemistry has greatly facilitated the development of prodrug strategies, enabling their activation through chemical and physical stimuli. This "on-demand" activation using bioorthogonal chemistry has revolutionized the research and development of prodrugs. Consequently, prodrug activation has garnered significant attention and emerged as an exciting field of translational research. This review summarizes the latest advancements in prodrug activation by utilizing bioorthogonal chemistry and mainly focuses on the activation of small-molecule prodrugs and antibody-drug conjugates. In addition, this review also discusses the opportunities and challenges of translating these advancements into clinical practice.
Collapse
Affiliation(s)
- Qunfeng Fu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
| | - Siyong Shen
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Pengwei Sun
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhi Gu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Yifei Bai
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Xianglin Wang
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhibo Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
- Peking University-Tsinghua University Center for Life Sciences, Peking University, Beijing 100871, China
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
7
|
Yang S, Wang L, Loredo A, Wang S, Ada N, Xiao H. Visible light-activated prodrug system with a novel heavy-atom-free photosensitizer. Bioorg Med Chem Lett 2023; 91:129365. [PMID: 37290494 PMCID: PMC11298069 DOI: 10.1016/j.bmcl.2023.129365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/20/2023] [Accepted: 06/02/2023] [Indexed: 06/10/2023]
Abstract
The use of light to activate prodrugs offers a promising method for the precise control of drug release, reducing drug-related side effects, and enhancing therapeutic effectiveness. We have created a novel prodrug system that utilizes a unique, heavy-atom-free photosensitizer to produce singlet oxygen, which then triggers the conversion of the prodrug into its active form. This system has been successfully demonstrated through the creation of "photo-unclick" prodrugs of paclitaxel (PTX), combretastatin A-4 (CA-4), and 10-hydroxy-7-ethylcamptothecin (SN-38). These prodrugs show decreased toxicity in the absence of light, but exhibit increased toxicity when exposed to red light.
Collapse
Affiliation(s)
- Shudan Yang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States
| | - Lushun Wang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States
| | - Axel Loredo
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States
| | - Shichao Wang
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States
| | - Nischal Ada
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States
| | - Han Xiao
- Department of Chemistry, Rice University, 6100 Main Street, Houston, TX 77005, United States; Department of Biosciences, Rice University, 6100 Main Street, Houston, TX 77005, United States; Department of Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, United States.
| |
Collapse
|
8
|
Zhang P, Zhu Y, Xiao C, Chen X. Activatable dual-functional molecular agents for imaging-guided cancer therapy. Adv Drug Deliv Rev 2023; 195:114725. [PMID: 36754284 DOI: 10.1016/j.addr.2023.114725] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 01/16/2023] [Accepted: 02/02/2023] [Indexed: 02/08/2023]
Abstract
Theranostics has attracted great attention due to its ability to combine the real-time diagnosis of cancers with efficient treatment modalities. Activatable dual-functional molecular agents could be synthesized by covalently conjugating imaging agents, therapeutic agents, stimuli-responsive linkers and/or targeting molecules together. They could be selectively activated by overexpressed physiological stimuli or external triggers at the tumor sites to release imaging agents and cytotoxic drugs, thus offering many advantages for tumor imaging and therapy, such as a high signal-to-noise ratio, low systemic toxicity, and improved therapeutic effects. This review summarizes the recent advances of dual-functional molecular agents that respond to various physiological or external stimuli for cancer theranostics. The molecular designs, synthetic strategies, activatable mechanisms, and biomedical applications of these molecular agents are elaborated, followed by a brief discussion of the challenges and opportunities in this field.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China; State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200433, PR China
| | - Yaowei Zhu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| |
Collapse
|
9
|
Jia S, Sletten EM. Spatiotemporal Control of Biology: Synthetic Photochemistry Toolbox with Far-Red and Near-Infrared Light. ACS Chem Biol 2022; 17:3255-3269. [PMID: 34516095 PMCID: PMC8918031 DOI: 10.1021/acschembio.1c00518] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The complex network of naturally occurring biological pathways motivates the development of new synthetic molecules to perturb and/or detect these processes for fundamental research and clinical applications. In this context, photochemical tools have emerged as an approach to control the activity of drug or probe molecules at high temporal and spatial resolutions. Traditional photochemical tools, particularly photolabile protecting groups (photocages) and photoswitches, rely on high-energy UV light that is only applicable to cells or transparent model animals. More recently, such designs have evolved into the visible and near-infrared regions with deeper tissue penetration, enabling photocontrol to study biology in tissue and model animal contexts. This Review highlights recent developments in synthetic far-red and near-infrared photocages and photoswitches and their current and potential applications at the interface of chemistry and biology.
Collapse
Affiliation(s)
- Shang Jia
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| | - Ellen M Sletten
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
10
|
Development of novel porphyrin/combretastatin A-4 conjugates for bimodal chemo and photodynamic therapy: Synthesis, photophysical and TDDFT computational studies. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.114138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
11
|
Guo K, Ma X, Li J, Zhang C, Wu L. Recent advances in combretastatin A-4 codrugs for cancer therapy. Eur J Med Chem 2022; 241:114660. [PMID: 35964428 DOI: 10.1016/j.ejmech.2022.114660] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
CA4 is a potent microtubule polymerization inhibitor and vascular disrupting agent. However, the in vivo efficiency of CA4 is limited owing to its poor pharmacokinetics resulting from its high lipophilicity and low water solubility. To improve the water solubility, CA4 phosphate (CA4P) has been developed and shows potent antivascular and antitumor effects. CA4P had been evaluated as a vascular disrupting agent in previousc linical trials. However, it had been discontinued due to the lack of a meaningful improvement in progression-free survival and unfavorable partial response data. Codrug is a drug design approach to chemically bind two or more drugs to improve therapeutic efficiency or decrease adverse effects. This review describes the progress made over the last twenty years in developing CA4-based codrugs to improve the therapeutic profile and achieve targeted delivery to cancer tissues. It also discusses the existing problems and the developmental prospects of CA4 codrugs.
Collapse
Affiliation(s)
- Kerong Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Ma
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jian Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chong Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China
| | - Liqiang Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
12
|
Rennie CC, Edkins RM. Targeted cancer phototherapy using phthalocyanine-anticancer drug conjugates. Dalton Trans 2022; 51:13157-13175. [PMID: 36018269 DOI: 10.1039/d2dt02040h] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Phototherapy, the use of light to selectively ablate cancerous tissue, is a compelling prospect. Phototherapy is divided into two major domains: photodynamic and photothermal, whereby photosensitizer irradiation generates reactive oxygen species or heat, respectively, to disrupt the cancer microenvironment. Phthalocyanines (Pcs) are prominent phototherapeutics due to their desirable optical properties and structural versatility. Targeting of Pc photosensitizers historically relied on the enhanced permeation and retention effect, but the weak specificity engendered by this approach has hindered bench-to-clinic translation. To improve specificity, antibody and peptide active-targeting groups have been employed to some effect. An alternative targeting method exploits the binding of anticancer drugs to direct the photosensitizer close to essential cellular components, allowing for precise, synergistic phototherapy. This Perspective explores the use of Pc-drug conjugates as targeted anticancer phototherapeutic systems with examples of Pc-platin, Pc-kinase, and Pc-anthracycline conjugates discussed in detail.
Collapse
Affiliation(s)
- Christopher C Rennie
- WestCHEM Department of Pure and Applied Chemistry, Thomas Graham Building, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| | - Robert M Edkins
- WestCHEM Department of Pure and Applied Chemistry, Thomas Graham Building, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| |
Collapse
|
13
|
Advantages of combined photodynamic therapy in the treatment of oncological diseases. Biophys Rev 2022; 14:941-963. [DOI: 10.1007/s12551-022-00962-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/24/2022] [Indexed: 12/22/2022] Open
|
14
|
de las Heras E, Sagristá ML, Agut M, Nonell S. Photosensitive EGFR-Targeted Nanocarriers for Combined Photodynamic and Local Chemotherapy. Pharmaceutics 2022; 14:pharmaceutics14020405. [PMID: 35214137 PMCID: PMC8880051 DOI: 10.3390/pharmaceutics14020405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/30/2022] [Accepted: 02/04/2022] [Indexed: 02/01/2023] Open
Abstract
The major limitation of any cancer therapy lies in the difficulty of precisely controlling the localization of the drug in the tumor cells. To improve this drawback, our study explores the use of actively-targeted chemo-photo-nanocarriers that recognize and bind to epidermal growth factor receptor-overexpressing cells and promote the local on-demand release of the chemotherapeutic agent doxorubicin triggered by light. Our results show that the attachment of high concentrations of doxorubicin to cetuximab-IRDye700DX-mesoporous silica nanoparticles yields efficient and selective photokilling of EGFR-expressing cells mainly through singlet oxygen-induced release of the doxorubicin from the nanocarrier and without any dark toxicity. Therefore, this novel triply functionalized nanosystem is an effective and safe nanodevice for light-triggered on-demand doxorubicin release.
Collapse
Affiliation(s)
- Elena de las Heras
- Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.d.l.H.); (M.A.)
| | - M. Lluïsa Sagristá
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain;
| | - Montserrat Agut
- Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.d.l.H.); (M.A.)
| | - Santi Nonell
- Institut Químic de Sarrià, Universitat Ramon Llull, 08017 Barcelona, Spain; (E.d.l.H.); (M.A.)
- Correspondence:
| |
Collapse
|
15
|
Otvagin VF, Kuzmina NS, Kudriashova ES, Nyuchev AV, Gavryushin AE, Fedorov AY. Conjugates of Porphyrinoid-Based Photosensitizers with Cytotoxic Drugs: Current Progress and Future Directions toward Selective Photodynamic Therapy. J Med Chem 2022; 65:1695-1734. [DOI: 10.1021/acs.jmedchem.1c01953] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vasilii F. Otvagin
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Natalia S. Kuzmina
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Ekaterina S. Kudriashova
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | - Alexander V. Nyuchev
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| | | | - Alexey Yu. Fedorov
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russian Federation
| |
Collapse
|
16
|
Paidakula S, Nerella S, Kankala S, Kankala RK. Recent Trends in Tubulin-Binding Combretastatin A-4 Analogs for Anticancer Drug Development. Curr Med Chem 2021; 29:3748-3773. [PMID: 34856892 DOI: 10.2174/0929867328666211202101641] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
Abstract
Although significant progress over several decades has been evidenced in cancer therapy, there still remains a need for the development of novel and effective therapeutic strategies to treat several relapsed and intractable cancers. In this regard, tubulin protein has become one of the efficient and major targets for anticancer drug discovery. Considering the antimitotic ability, several tubulin inhibitors have been developed to act against various cancers. Among various tubulin inhibitors available, combretastatin-A4 (CA-4), a naturally occurring lead molecule, offers exceptional cytotoxicity (including the drug-resistant cell lines) and antivascular effects. Although CA-4 offers exceptional therapeutic efficacy, several new advancements have been proposed, such as the structural modification via A and B rings, as well as cis-olefinic bridging, which provide highly efficient analogs with improved tubulin-binding efficiency to meet the anticancer drug development requirements. This review systematically emphasizes the recent trends and latest developments in the anticancer drug design & discovery, using CA-4 analogs as the tubulin inhibiting agents, highlighting their structure-activity relationships (SAR) and resultant pharmacological efficacies.
Collapse
Affiliation(s)
- Suresh Paidakula
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Srinivas Nerella
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Shravankumar Kankala
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | | |
Collapse
|
17
|
Pham TC, Nguyen VN, Choi Y, Lee S, Yoon J. Recent Strategies to Develop Innovative Photosensitizers for Enhanced Photodynamic Therapy. Chem Rev 2021; 121:13454-13619. [PMID: 34582186 DOI: 10.1021/acs.chemrev.1c00381] [Citation(s) in RCA: 772] [Impact Index Per Article: 193.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This review presents a robust strategy to design photosensitizers (PSs) for various species. Photodynamic therapy (PDT) is a photochemical-based treatment approach that involves the use of light combined with a light-activated chemical, referred to as a PS. Attractively, PDT is one of the alternatives to conventional cancer treatment due to its noninvasive nature, high cure rates, and low side effects. PSs play an important factor in photoinduced reactive oxygen species (ROS) generation. Although the concept of photosensitizer-based photodynamic therapy has been widely adopted for clinical trials and bioimaging, until now, to our surprise, there has been no relevant review article on rational designs of organic PSs for PDT. Furthermore, most of published review articles in PDT focused on nanomaterials and nanotechnology based on traditional PSs. Therefore, this review aimed at reporting recent strategies to develop innovative organic photosensitizers for enhanced photodynamic therapy, with each example described in detail instead of providing only a general overview, as is typically done in previous reviews of PDT, to provide intuitive, vivid, and specific insights to the readers.
Collapse
Affiliation(s)
- Thanh Chung Pham
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| | - Yeonghwan Choi
- Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Songyi Lee
- Department of Chemistry, Pukyong National University, Busan 48513, Korea.,Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan 48513, Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
18
|
Yang DC, Wang S, Weng XL, Zhang HX, Liu JY, Lin Z. Singlet Oxygen-Responsive Polymeric Nanomedicine for Light-Controlled Drug Release and Image-Guided Photodynamic-Chemo Combination Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:33905-33914. [PMID: 34278780 DOI: 10.1021/acsami.1c09044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Coencapsulation of chemotherapeutic agents and photosensitizers into nanocarriers can help to achieve a combination of chemotherapy and photodynamic therapy for superior antitumor effects. However, precise on-demand drug release remains a major challenge. In addition, the loaded photosensitizers usually tend to aggregate, which can significantly weaken their fluorescent signals and photodynamic activities. To address these issues, herein, a smart nanocarrier termed as singlet oxygen-responsive nanoparticle (SOR-NP) was constructed by introducing singlet oxygen (1O2)-sensitive aminoacrylate linkers into amphiphilic mPEG-b-PCL copolymers. Boron dipyrromethene (BDP) and paclitaxel (PTX) as model therapeutic agents were coloaded into an 1O2-responsive nanocarrier for realizing light-controlled drug release and combination cancer treatment. This polymeric nanocarrier could substantially relieve the aggregation of encapsulated BDP due to the presence of a long hydrophobic chain. Therefore, the formed SOR-NPBDP/PTX nanodrug could generate bright fluorescent signals and high levels of 1O2, which could mediate cell death via PDT and rupture aminoacrylate linker simultaneously, leading to collapse of SOR-NPBDP/PTX and subsequent PTX release. The light-triggered drug release and combined anticancer effects of SOR-NPBDP/PTX were validated in HepG2 and MCF-7 cancer cells and H22 tumor-bearing mice. This study provides a promising strategy for tumor-specific drug release and selective photodynamic-chemo combination treatment.
Collapse
Affiliation(s)
- De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Shuai Wang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Xiao-Lu Weng
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Hong-Xia Zhang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou 350108, China
- State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Zhonghui Lin
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
19
|
Xu G, Zhang HX, Li XQ, Yang DC, Liu JY. Red light triggered photodynamic-chemo combination therapy using a prodrug caged by photosensitizer. Eur J Med Chem 2021; 215:113251. [PMID: 33611187 DOI: 10.1016/j.ejmech.2021.113251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/10/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022]
Abstract
Development of the drug with high therapeutic efficacy and low toxicity is crucial to cancer ablation. In this study, we have demonstrated a red light-responsive prodrug BDP-TK-CPT by connecting the chemotherapeutic agent camptothecin with a boron dipyrromethene (BDP)-based photosensitizer via a reactive oxygen species (ROS)-labile thioketal chain. Since camptothecin is modified by a BDP-based macrocycle at the active site, the formed prodrug displays an extremely low toxicity in dark. However, upon illumination by red light, it can efficiently generate ROS leading to cell death by photodynamic therapy. Meanwhile, the ROS generated can destroy thioketal group to release free camptothecin which further results in local cell death by chemotherapy. The combined antitumor effects of the prodrug have been verified in HepG2, EC109, and HeLa cancer cells and mice bearing H22 tumors. This study may provide an alternative strategy for stimuli-responsive combination treatment of tumors by conjugation of ROS-activatable prodrugs with photosensitizing agents.
Collapse
Affiliation(s)
- Gan Xu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Hong-Xia Zhang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiao-Qiang Li
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies & Key Laboratory of Molecule Synthesis and Function Discovery (Fujian Province University), College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
20
|
Zhao X, Liu J, Fan J, Chao H, Peng X. Recent progress in photosensitizers for overcoming the challenges of photodynamic therapy: from molecular design to application. Chem Soc Rev 2021; 50:4185-4219. [PMID: 33527104 DOI: 10.1039/d0cs00173b] [Citation(s) in RCA: 561] [Impact Index Per Article: 140.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Photodynamic therapy (PDT), a therapeutic mode involving light triggering, has been recognized as an attractive oncotherapy treatment. However, nonnegligible challenges remain for its further clinical use, including finite tumor suppression, poor tumor targeting, and limited therapeutic depth. The photosensitizer (PS), being the most important element of PDT, plays a decisive role in PDT treatment. This review summarizes recent progress made in the development of PSs for overcoming the above challenges. This progress has included PSs developed to display enhanced tolerance of the tumor microenvironment, improved tumor-specific selectivity, and feasibility of use in deep tissue. Based on their molecular photophysical properties and design directions, the PSs are classified by parent structures, which are discussed in detail from the molecular design to application. Finally, a brief summary of current strategies for designing PSs and future perspectives are also presented. We expect the information provided in this review to spur the further design of PSs and the clinical development of PDT-mediated cancer treatments.
Collapse
Affiliation(s)
- Xueze Zhao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, 116024 Dalian, China.
| | | | | | | | | |
Collapse
|
21
|
Mitra K, Hartman MCT. Silicon phthalocyanines: synthesis and resurgent applications. Org Biomol Chem 2021; 19:1168-1190. [DOI: 10.1039/d0ob02299c] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Their unique axial bonds and NIR optical properties have made silicon phthalocyanines (SiPcs) valuable compounds. Herein, we present key synthetic strategies and emerging applications of SiPcs over the past decade.
Collapse
Affiliation(s)
- Koushambi Mitra
- Department of Chemistry
- Virginia Commonwealth University
- Richmond
- USA
- Massey Cancer Center
| | - Matthew C. T. Hartman
- Department of Chemistry
- Virginia Commonwealth University
- Richmond
- USA
- Massey Cancer Center
| |
Collapse
|
22
|
Weinstain R, Slanina T, Kand D, Klán P. Visible-to-NIR-Light Activated Release: From Small Molecules to Nanomaterials. Chem Rev 2020; 120:13135-13272. [PMID: 33125209 PMCID: PMC7833475 DOI: 10.1021/acs.chemrev.0c00663] [Citation(s) in RCA: 324] [Impact Index Per Article: 64.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Indexed: 02/08/2023]
Abstract
Photoactivatable (alternatively, photoremovable, photoreleasable, or photocleavable) protecting groups (PPGs), also known as caged or photocaged compounds, are used to enable non-invasive spatiotemporal photochemical control over the release of species of interest. Recent years have seen the development of PPGs activatable by biologically and chemically benign visible and near-infrared (NIR) light. These long-wavelength-absorbing moieties expand the applicability of this powerful method and its accessibility to non-specialist users. This review comprehensively covers organic and transition metal-containing photoactivatable compounds (complexes) that absorb in the visible- and NIR-range to release various leaving groups and gasotransmitters (carbon monoxide, nitric oxide, and hydrogen sulfide). The text also covers visible- and NIR-light-induced photosensitized release using molecular sensitizers, quantum dots, and upconversion and second-harmonic nanoparticles, as well as release via photodynamic (photooxygenation by singlet oxygen) and photothermal effects. Release from photoactivatable polymers, micelles, vesicles, and photoswitches, along with the related emerging field of photopharmacology, is discussed at the end of the review.
Collapse
Affiliation(s)
- Roy Weinstain
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Tomáš Slanina
- Institute
of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nám. 2, 166 10 Prague, Czech Republic
| | - Dnyaneshwar Kand
- School
of Plant Sciences and Food Security, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Petr Klán
- Department
of Chemistry and RECETOX, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
23
|
Zhao C, Chen J, Zhong R, Chen DS, Shi J, Song J. Materialien mit Selektivität für oxidative Molekülspezies für die Diagnostik und Therapie. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201915833] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Caiyan Zhao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jingxiao Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Pharmaceutical Sciences Jiangnan University Wuxi 214122 PR China
| | - Ruibo Zhong
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Dean Shuailin Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jinjun Shi
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
| |
Collapse
|
24
|
Zhao C, Chen J, Zhong R, Chen DS, Shi J, Song J. Oxidative‐Species‐Selective Materials for Diagnostic and Therapeutic Applications. Angew Chem Int Ed Engl 2020; 60:9804-9827. [DOI: 10.1002/anie.201915833] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/15/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Caiyan Zhao
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jingxiao Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
- Key Laboratory of Carbohydrate Chemistry and Biotechnology Ministry of Education School of Pharmaceutical Sciences Jiangnan University Wuxi 214122 PR China
| | - Ruibo Zhong
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Dean Shuailin Chen
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jinjun Shi
- Center for Nanomedicine Brigham and Women's Hospital Harvard Medical School Boston Massachusetts 02115 USA
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and Biology Institution College of Chemistry Fuzhou University Fuzhou 350108 China
| |
Collapse
|
25
|
Ha SYY, Zhou Y, Fong WP, Ng DKP. Multifunctional Molecular Therapeutic Agent for Targeted and Controlled Dual Chemo- and Photodynamic Therapy. J Med Chem 2020; 63:8512-8523. [DOI: 10.1021/acs.jmedchem.0c00893] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
26
|
Watanabe K, Terao N, Kii I, Nakagawa R, Niwa T, Hosoya T. Indolizines Enabling Rapid Uncaging of Alcohols and Carboxylic Acids by Red Light-Induced Photooxidation. Org Lett 2020; 22:5434-5438. [DOI: 10.1021/acs.orglett.0c01799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kenji Watanabe
- Laboratory for Chemical Biology, RIKEN Center for Biosystems Dynamics Research (BDR), 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Nodoka Terao
- Laboratory for Chemical Biology, RIKEN Center for Biosystems Dynamics Research (BDR), 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Isao Kii
- Laboratory for Chemical Biology, RIKEN Center for Biosystems Dynamics Research (BDR), 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
- RIKEN Cluster for Science, Technology and Innovation Hub, 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
- Laboratory for Drug Target Research, Integrated Bioscience Division, Institute of Agriculture, Shinshu University, 8304 minami-Minowa, Kami-Ina, Nagano, 399-4598, Japan
| | - Reiko Nakagawa
- Laboratory for Phyloinformatics, RIKEN Center for Biosystems Dynamics Research (BDR), 2-2-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takashi Niwa
- Laboratory for Chemical Biology, RIKEN Center for Biosystems Dynamics Research (BDR), 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takamitsu Hosoya
- Laboratory for Chemical Biology, RIKEN Center for Biosystems Dynamics Research (BDR), 6-7-3 minatojima-minamimachi, Chuo-ku, Kobe 650-0047, Japan
- Laboratory of Chemical Bioscience, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
27
|
Luciano MP, Nourian S, Gorka AP, Nani RR, Nagaya T, Kobayashi H, Schnermann MJ. A near-infrared light-mediated cleavable linker strategy using the heptamethine cyanine chromophore. Methods Enzymol 2020; 641:245-275. [PMID: 32713525 PMCID: PMC10763689 DOI: 10.1016/bs.mie.2020.04.043] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Optical methods offer the potential to manipulate living biological systems with exceptional spatial and temporal control. Caging bioactive molecules with photocleavable functional groups is an important strategy that could be applied to a range of problems, including the targeted delivery of otherwise toxic therapeutics. However existing approaches that require UV or blue light are difficult to apply in organismal settings due to issues of tissue penetration and light toxicity. Photocaging groups built on the heptamethine cyanine scaffold enable the targeted delivery of bioactive molecules using near-IR light (up to 780nm) in live animal settings. Here we provide a detailed procedure demonstrating the utility of the heptamethine cyanine caging group to create a light-cleavable linker between an antibody, panitumumab, and a therapeutic small molecule in the duocarmycin class of natural products. Descriptions of the design and synthesis of the small molecule component, assembly of the antibody conjugate, in vitro analysis of uncaging, in vivo imaging, and impact on tumor progression are provided.
Collapse
Affiliation(s)
- Michael P Luciano
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Saghar Nourian
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Alexander P Gorka
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Roger R Nani
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States
| | - Tadanobu Nagaya
- Laboratory of Molecular Theranostics, NIH/NCI/CCR, Bethesda, MD, United States
| | - Hisataka Kobayashi
- Laboratory of Molecular Theranostics, NIH/NCI/CCR, Bethesda, MD, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, United States.
| |
Collapse
|
28
|
Subramaniyan B, Rajaputra P, Nguyen L, Li M, Peer CJ, Kindrick J, Figg WD, Woo S, You Y. Local and Systemic Antitumor Effects of Photo-activatable Paclitaxel Prodrug on Rat Breast Tumor Models. Photochem Photobiol 2020; 96:668-679. [PMID: 31883393 PMCID: PMC8043141 DOI: 10.1111/php.13202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
We demonstrated that a large primary and a small untreated distant breast cancer could be controlled by local treatment with our light-activatable paclitaxel (PTX) prodrug. We hypothesized that the treated tumor would be damaged by the combinational effects of photodynamic therapy (PDT) and locally released PTX and that the distant tumor would be suppressed by systemic antitumor effects. Syngeneic rat breast cancer models (single- and two-tumor models) were established on Fischer 344 rats by subcutaneous injection of MAT B III cells. The rats were injected with PTX prodrug (dose: 1 umole kg-1 , i.v.), and tumors were treated with illumination using a 690-nm laser (75 or 140 mW cm-1 for 30 min, cylindrical light diffuser, drug-light interval [DLI] 9 h). Larger tumors (~16 mm) were effectively ablated (100%) without recurrence for >90 days. All cured rats rejected rechallenged tumor for up to 12 months. In the two-tumor model, the treatment of the local large tumor (~16 mm) also cured the untreated tumor (4-6 mm) through adaptive immune activation. This is our first demonstration that local treatment with our PTX prodrug produces systemic antitumor effects. Further investigations are warranted to understand mechanisms and optimal conditions to achieve clinically translatable systemic antitumor effects.
Collapse
Affiliation(s)
- Bharathiraja Subramaniyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Pallavi Rajaputra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Luong Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - Jessica Kindrick
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Youngjae You
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
29
|
Nguyen L, Li M, Woo S, You Y. Development of Prodrugs for PDT-Based Combination Therapy Using a Singlet-Oxygen-Sensitive Linker and Quantitative Systems Pharmacology. J Clin Med 2019; 8:jcm8122198. [PMID: 31847080 PMCID: PMC6947033 DOI: 10.3390/jcm8122198] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Photodynamic therapy (PDT) has become an effective treatment for certain types of solid tumors. The combination of PDT with other therapies has been extensively investigated in recent years to improve its effectiveness and expand its applications. This focused review summarizes the development of a prodrug system in which anticancer drugs are activated locally at tumor sites during PDT treatment. The development of a singlet-oxygen-sensitive linker that can be conveniently conjugated to various drugs and efficiently cleaved to release intact drugs is recapitulated. The initial design of prodrugs, preliminary efficacy evaluation, pharmacokinetics study, and optimization using quantitative systems pharmacology is discussed. Current treatment optimization in animal models using physiologically based a pharmacokinetic (PBPK) modeling approach is also explored.
Collapse
Affiliation(s)
- Luong Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
| | - Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA; (L.N.); (M.L.); (S.W.)
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14214, USA
- Correspondence: ; Tel.: +1-716-645-4843
| |
Collapse
|
30
|
Peiró Cadahía J, Previtali V, Troelsen NS, Clausen MH. Prodrug strategies for targeted therapy triggered by reactive oxygen species. MEDCHEMCOMM 2019; 10:1531-1549. [PMID: 31673314 PMCID: PMC6786010 DOI: 10.1039/c9md00169g] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Abstract
Increased levels of reactive oxygen species (ROS) have been associated with numerous pathophysiological conditions including cancer and inflammation and the ROS stimulus constitutes a potential trigger for drug delivery strategies. Over the past decade, a number of ROS-sensitive functionalities have been identified with the purpose of introducing disease-targeting properties into small molecule drugs - a prodrug strategy that offers a promising approach for increasing the selectivity and efficacy of treatments. This review will provide an overview of the ROS-responsive prodrugs developed to date. A discussion on the current progress and limitations is provided along with a reflection on the unanswered questions that need to be addressed in order to advance this novel approach to the clinic.
Collapse
Affiliation(s)
| | - Viola Previtali
- Center for Nanomedicine & Theranostics , Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK 2800 , Kongens Lyngby , Denmark .
| | - Nikolaj S Troelsen
- Center for Nanomedicine & Theranostics , Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK 2800 , Kongens Lyngby , Denmark .
| | - Mads H Clausen
- Center for Nanomedicine & Theranostics , Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , DK 2800 , Kongens Lyngby , Denmark .
| |
Collapse
|
31
|
Li M, Nguyen L, Subramaniyan B, Bio M, Peer CJ, Kindrick J, Figg WD, Woo S, You Y. PBPK modeling-based optimization of site-specific chemo-photodynamic therapy with far-red light-activatable paclitaxel prodrug. J Control Release 2019; 308:86-97. [PMID: 31299262 DOI: 10.1016/j.jconrel.2019.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
Photodynamic therapy (PDT) is a clinically approved therapeutic modality to treat certain types of cancers. However, incomplete ablation of tumor is a challenge. Visible and near IR-activatable prodrug, exhibiting the combined effects of PDT and local chemotherapy, showed better efficacy than PDT alone, without systemic side effects. Site-specifically released chemotherapeutic drugs killed cancer cells surviving from rapid PDT damage via bystander effects. Recently, we developed such a paclitaxel (PTX) prodrug that targets folate receptors. The goals of this study were to determine the optimal treatment conditions, based on modeling, for maximum antitumor efficacy in terms of drug-light interval (DLI), and to investigate the impact of rapid PDT effects on the pharmacokinetic (PK) profiles of the released PTX. PK profiles of the prodrug were determined in key organs and a quantitative systems pharmacology (QSP) model was established to simulate PK profiles of the prodrug and the released PTX. Three illumination time points (DLI = 0.5, 9, or 48 h) were selected for the treatment based on the plasma/tumor ratio of the prodrug to achieve V-PDT (vascular targeted-PDT, 0.5 h), C-PDT (cellular targeted-PDT, 48 h), or both V- and C-PDT (9 h). The anti-tumor efficacy of the PTX prodrug was greatly influenced by the DLI. The 9 h DLI group, when both tumor and plasma concentrations of the prodrug were sufficient, showed the best antitumor effect. The clearance of the released PTX from tumor seemed to be largely impacted by blood circulation. Here, QSP modeling was an invaluable tool for rational optimization of the treatment conditions and for a deeper mechanistic understanding of the positive physiological effect of the combination therapy.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA
| | - Luong Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA
| | - Bharathiraja Subramaniyan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA
| | - Moses Bio
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA
| | - Cody J Peer
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda 20892, MD, USA
| | - Jessica Kindrick
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda 20892, MD, USA
| | - William D Figg
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda 20892, MD, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA.
| | - Youngjae You
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City 73117, OK, USA.
| |
Collapse
|
32
|
Azodyes as markers for tumor hypoxia imaging and therapy: An up-to-date review. Chem Biol Interact 2019; 307:91-104. [DOI: 10.1016/j.cbi.2019.04.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/21/2019] [Accepted: 04/28/2019] [Indexed: 12/20/2022]
|
33
|
Hu X, Ogawa K, Li S, Kiwada T, Odani A. A Platinum Functional Porphyrin Conjugate: An Excellent Cancer Killer for Photodynamic Therapy. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2019. [DOI: 10.1246/bcsj.20180382] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xiaojun Hu
- Laboratory of Biosensing Technology, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China
- Division of Pharmaceutical Sciences, Graduate School of Medical Science, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Siqiaozhi Li
- Division of Pharmaceutical Sciences, Graduate School of Medical Science, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Tatsuto Kiwada
- College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Akira Odani
- College of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
34
|
Luby BM, Walsh CD, Zheng G. Advanced Photosensitizer Activation Strategies for Smarter Photodynamic Therapy Beacons. Angew Chem Int Ed Engl 2019; 58:2558-2569. [DOI: 10.1002/anie.201805246] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/08/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Benjamin M. Luby
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
| | - Connor D. Walsh
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
- Department of Medical BiophysicsUniversity of Toronto Toronto Ontario Canada
| |
Collapse
|
35
|
|
36
|
Saeed M, Iqbal MZ, Ren W, Xia Y, Khan WS, Wu A. Tunable fabrication of new theranostic Fe3O4-black TiO2 nanocomposites: dual wavelength stimulated synergistic imaging-guided phototherapy in cancer. J Mater Chem B 2019; 7:210-223. [DOI: 10.1039/c8tb02704h] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The development of a simplified theranostic system with high-efficiency for multifunctional imaging-guided photodynamic therapy/photothermal therapy (PDT/PTT) is a great challenge.
Collapse
Affiliation(s)
- Madiha Saeed
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - M. Zubair Iqbal
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Wenzhi Ren
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Yuanzhi Xia
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Waheed S. Khan
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Aiguo Wu
- CAS Key Laboratory of Magnetic Materials and Devices
- Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
37
|
Luby BM, Walsh CD, Zheng G. Advanced Photosensitizer Activation Strategies for Smarter Photodynamic Therapy Beacons. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201805246] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Benjamin M. Luby
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
| | - Connor D. Walsh
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
| | - Gang Zheng
- Princess Margaret Cancer Centre and Techna InstituteUniversity Health Network 101 College St. Toronto ON Canada
- Institute of Biomaterials and Biomedical EngineeringUniversity of Toronto Toronto Ontario Canada
- Department of Medical BiophysicsUniversity of Toronto Toronto Ontario Canada
| |
Collapse
|
38
|
Callaghan S, Senge MO. The good, the bad, and the ugly - controlling singlet oxygen through design of photosensitizers and delivery systems for photodynamic therapy. Photochem Photobiol Sci 2018; 17:1490-1514. [PMID: 29569665 DOI: 10.1039/c8pp00008e] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Singlet oxygen, although integral to photodynamic therapy, is notoriously uncontrollable, suffers from poor selectivity and has fast decomposition rates in biological media. Across the scientific community, there is a conscious effort to refine singlet oxygen interactions and initiate selective and controlled release to produce a consistent and reproducible therapeutic effect in target tissue. This perspective aims to provide an insight into the contemporary design principles behind photosensitizers and drug delivery systems that depend on a singlet oxygen response or controlled release. The discussion will be accompanied by in vitro and in vivo examples, in an attempt to highlight advancements in the field and future prospects for the more widespread application of photodynamic therapy.
Collapse
Affiliation(s)
- Susan Callaghan
- School of Chemistry, SFI Tetrapyrrole Laboratory, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mathias O Senge
- School of Chemistry, SFI Tetrapyrrole Laboratory, Trinity Biomedical Sciences Institute, Trinity College Dublin, the University of Dublin, 152-160 Pearse Street, Dublin 2, Ireland and Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
39
|
Four organic crystals displaying distinctively different emission colors based on an ESIPT-active organic molecule. CHINESE CHEM LETT 2018. [DOI: 10.1016/j.cclet.2018.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
40
|
Ling X, Zhang S, Liu Y, Bai M. Light-activatable cannabinoid prodrug for combined and target-specific photodynamic and cannabinoid therapy. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-9. [PMID: 30334393 DOI: 10.1117/1.jbo.23.10.108001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 09/19/2018] [Indexed: 06/08/2023]
Abstract
Cannabinoids are emerging as promising antitumor drugs. However, complete tumor eradication solely by cannabinoid therapy remains challenging. In this study, we developed a far-red light activatable cannabinoid prodrug, which allows for tumor-specific and combinatory cannabinoid and photodynamic therapy. This prodrug consists of a phthalocyanine photosensitizer (PS), reactive oxygen species (ROS)-sensitive linker, and cannabinoid. It targets the type-2 cannabinoid receptor (CB2R) overexpressed in various types of cancers. Upon the 690-nm light irradiation, the PS produces cytotoxic ROS, which simultaneously cleaves the ROS-sensitive linker and subsequently releases the cannabinoid drug. We found that this unique multifunctional prodrug design offered dramatically improved therapeutic efficacy, and therefore provided a new strategy for targeted, controlled, and effective antitumor cannabinoid therapy.
Collapse
Affiliation(s)
- Xiaoxi Ling
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
| | - Shaojuan Zhang
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
| | - Yang Liu
- Vanderbilt University Institute of Imaging Sciences, Nashville, Tennessee, United States
| | - Mingfeng Bai
- University of Pittsburgh, Department of Radiology, Pittsburgh, Pennsylvania, United States
- University of Pittsburgh, Department of Medicine, Pittsburgh Pennsylvania, United States
- University of Pittsburgh, Department of Bioengineering, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
41
|
Ghosh G, Belh SJ, Chiemezie C, Walalawela N, Ghogare AA, Vignoni M, Thomas AH, McFarland SA, Greer EM, Greer A. S,S-Chiral Linker Induced U Shape with a Syn-facial Sensitizer and Photocleavable Ethene Group. Photochem Photobiol 2018; 95:293-305. [PMID: 30113068 DOI: 10.1111/php.13000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 08/06/2018] [Indexed: 11/30/2022]
Abstract
There is a major need for light-activated materials for the release of sensitizers and drugs. Considering the success of chiral columns for the separation of enantiomer drugs, we synthesized an S,S-chiral linker system covalently attached to silica with a sensitizer ethene near the silica surface. First, the silica surface was modified to be aromatic rich, by replacing 70% of the surface groups with (3-phenoxypropyl)silane. We then synthesized a 3-component conjugate [chlorin sensitizer, S,S-chiral cyclohexane and ethene building blocks] in 5 steps with a 13% yield, and covalently bound the conjugate to the (3-phenoxypropyl)silane-coated silica surface. We hypothesized that the chiral linker would increase exposure of the ethene site for enhanced 1 O2 -based sensitizer release. However, the chiral linker caused the sensitizer conjugate to adopt a U shape due to favored 1,2-diaxial substituent orientation; resulting in a reduced efficiency of surface loading. Further accentuating the U shape was π-π stacking between the (3-phenoxypropyl)silane and sensitizer. Semiempirical calculations and singlet oxygen luminescence data provided deeper insight into the sensitizer's orientation and release. This study has lead to insight on modifications of surfaces for drug photorelease and can help lead to the development of miniaturized photodynamic devices.
Collapse
Affiliation(s)
- Goutam Ghosh
- Department of Chemistry, Acadia University, Wolfville, NS, Canada.,Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Sarah J Belh
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Callistus Chiemezie
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Niluksha Walalawela
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Ashwini A Ghogare
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| | - Mariana Vignoni
- INIFTA, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, CCT La Plata-CONICET, La Plata, Argentina
| | - Andrés H Thomas
- INIFTA, Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, CCT La Plata-CONICET, La Plata, Argentina
| | - Sherri A McFarland
- Department of Chemistry, Acadia University, Wolfville, NS, Canada.,Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC
| | - Edyta M Greer
- Department of Natural Sciences, Baruch College of the City University of New York, New York, NY
| | - Alexander Greer
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, NY.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY
| |
Collapse
|
42
|
Hou Y, Zhou Z, Huang K, Yang H, Han G. Long Wavelength Light Activated Prodrug Conjugates for Biomedical Applications. CHEMPHOTOCHEM 2018. [DOI: 10.1002/cptc.201800147] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Yutong Hou
- The Key Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and SensorsShanghai Normal University Shanghai 200234 China
| | - Zhiguo Zhou
- The Key Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and SensorsShanghai Normal University Shanghai 200234 China
| | - Kai Huang
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School Worcester Massachusetts 01605 United States
| | - Hong Yang
- The Key Laboratory of Resource Chemistry of Ministry of Education, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and SensorsShanghai Normal University Shanghai 200234 China
| | - Gang Han
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School Worcester Massachusetts 01605 United States
| |
Collapse
|
43
|
Liu W, Liu H, Peng X, Zhou G, Liu D, Li S, Zhang J, Wang S. Hypoxia-Activated Anticancer Prodrug for Bioimaging, Tracking Drug Release, and Anticancer Application. Bioconjug Chem 2018; 29:3332-3343. [PMID: 30192132 DOI: 10.1021/acs.bioconjchem.8b00511] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel anticancer theranostic prodrug, FDU-DB-NO2, specifically activated by hypoxia for selective two-photon imaging hypoxia status, real-time tracking drug release, and solid tumor therapy was designed. The devised prodrug consists of an anticancer drug floxuridine (FDU), a fluorescence dye precursor 4'-(diethylamino)-1,1'-biphenyl-2-carboxylate (DB), and a hypoxic trigger 4-nitrobenzyl group. In normal cells, FDU-DB-NO2 is "locked". Whereas in tumor cells, the prodrug is "unlocked" by hypoxia and results in fluorescent dye 7-(diethylamino)coumarin (CM) generation along with FDU release. The amounts and rates of CM formation and FDU release were controlled by hypoxic status and increased with the decreasing of the O2 concentration. The hypoxic status, distribution of oxygen, and amount of FDU release in tumor cells, spheroids, and tumor tissue could be visualized by fluorescence. FDU-DB-NO2 showed high cytotoxicity against hypoxic MCF-7 and MCG-803 cell lines and no cytotoxicity against normoxic BRL-3A cells and exhibited effective inhibition on tumor growth of MCF-7-cell-inoculated xenograft nude mice. This strategy may provide a promising platform for selective two-photon imaging hypoxia status, real-time tracking drug release, and personalized solid tumor treatment.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Haitong Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Xiaoran Peng
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Guoqiang Zhou
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Dandan Liu
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Shenghui Li
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Jinchao Zhang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| | - Shuxiang Wang
- Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Key Laboratory of Chemical Biology of Hebei Province, College of Chemistry and Environmental Science , Hebei University , Baoding 071002 , China
| |
Collapse
|
44
|
Anderson ED, Sova S, Ivanic J, Kelly L, Schnermann MJ. Defining the conditional basis of silicon phthalocyanine near-IR ligand exchange. Phys Chem Chem Phys 2018; 20:19030-19036. [PMID: 29971294 PMCID: PMC6344126 DOI: 10.1039/c8cp03842b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bond cleavage reactions initiated by long-wavelength light are needed to extend the scope of the caged-uncaged paradigm into complex physiological settings. Axially unsymmetrical silicon phthalocyanines (SiPcs) undergo efficient release of phenol ligands in a reaction contingent on three factors - near-IR light (690 nm), hypoxia, and a thiol reductant. These studies detail efforts to define the mechanistic basis for this unique conditionally-dependent bond cleavage reaction. Spectroscopic studies provide evidence for the formation of a key phthalocyanine radical anion intermediate formed from the triplet state in a reductant-dependent manner. Computational chemistry studies indicate that phenol ligand solvolysis proceeds through a heptacoordinate silicon transition state and that this solvolytic process is favored following SiPc radical anion formation. These results provide insight regarding the central role that radical anion intermediates formed through photoinduced electron transfer with biological reductants can play in long-wavelength uncaging reactions.
Collapse
Affiliation(s)
- Erin D Anderson
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, USA.
| | | | | | | | | |
Collapse
|
45
|
|
46
|
Tao W, He Z. ROS-responsive drug delivery systems for biomedical applications. Asian J Pharm Sci 2018; 13:101-112. [PMID: 32104383 PMCID: PMC7032079 DOI: 10.1016/j.ajps.2017.11.002] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/28/2017] [Accepted: 11/28/2017] [Indexed: 02/06/2023] Open
Abstract
In the field of biomedicine, stimuli-responsive drug delivery systems (DDSs) have become increasingly popular due to their site-specific release ability in response to a certain physiological stimulus, which may result in both enhanced treatment outcome and reduced side effects. Reactive oxygen species (ROS) are the unavoidable consequence of cell oxidative metabolism. ROS play a crucial part in regulating biological and physiological processes, whereas excessive intracellular ROS usually lead to the oxidation stress which has implications in several typical diseases such as cancer, inflammation and atherosclerosis. Therefore, ROS-responsive DDSs have elicited widespread popularity for their promising applications in a series of biomedical research because the payload is only released in targeted cells or tissues that overproduce ROS. According to the design of ROS-responsive DDSs, the main release mechanisms of therapeutic agents can be ascribed to ROS-induced carrier solubility change, ROS-induced carrier cleavage or ROS-induced prodrug linker cleavage. This review summarized the latest development and novel design of ROS-responsive DDSs and discussed their design concepts and the applications in the biomedical field.
Collapse
Affiliation(s)
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang 110016, China
| |
Collapse
|
47
|
Zhang J, Jiang C, Figueiró Longo JP, Azevedo RB, Zhang H, Muehlmann LA. An updated overview on the development of new photosensitizers for anticancer photodynamic therapy. Acta Pharm Sin B 2018; 8:137-146. [PMID: 29719775 PMCID: PMC5925394 DOI: 10.1016/j.apsb.2017.09.003] [Citation(s) in RCA: 352] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/14/2017] [Accepted: 07/15/2017] [Indexed: 12/31/2022] Open
Abstract
Photodynamic therapy (PDT), based on the photoactivation of photosensitizers (PSs), has become a well-studied therapy for cancer. Photofrin®, belonging to the first generation of PS, is still widely used for the treatment of different kinds of cancers; however, it has several drawbacks that significantly limit its general clinical use. Consequently, there has been extensive research on the design of PS molecules with optimized pharmaceutical properties, with aiming of overcoming the disadvantages of traditional PS, such as poor chemical purity, long half-life, excessive accumulation into the skin, and low attenuation coefficients. The rational design of novel PS with desirable properties has attracted considerable research in the pharmaceutical field. This review presents an overview on the classical photosensitizers and the most significant recent advances in the development of PS with regard to their potential application in oncology.
Collapse
Affiliation(s)
- Juan Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
- Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil
| | - Chengshi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | | | | | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Luis Alexandre Muehlmann
- Institute of Biological Sciences, University of Brasilia, Brasilia 70910-900, Brazil
- Faculty of Ceilandia, University of Brasilia, Brasilia 72220-900, Brazil
| |
Collapse
|
48
|
Thapa P, Li M, Karki R, Bio M, Rajaputra P, Nkepang G, Woo S, You Y. Folate-PEG Conjugates of a Far-Red Light-Activatable Paclitaxel Prodrug to Improve Selectivity toward Folate Receptor-Positive Cancer Cells. ACS OMEGA 2017; 2:6349-6360. [PMID: 29104951 PMCID: PMC5664141 DOI: 10.1021/acsomega.7b01105] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 09/19/2017] [Indexed: 05/23/2023]
Abstract
We recently demonstrated the far-red light-activatable prodrug of paclitaxel (PTX), Pc-(L-PTX)2. Upon illumination with a 690 nm laser, Pc-(L-PTX)2 showed combinational cell killing from rapid photodynamic therapy damage by singlet oxygen, followed by sustained chemotherapy effects from locally released PTX. However, its high lipophilicity (log D7.4 > 3.1) caused aggregation in aqueous solutions and has nonselectivity toward cancer cells. To solve these important problems, we prepared folic acid (FA)-conjugated and photoactivatable prodrugs of PTX with a polyethylene glycol (PEG) spacer of various chain lengths: FA-PEG n -Pc-L-PTX [n = 0 (0k, 5), ∼23 (1k, 7a), ∼45 (2k, 7b), ∼80 (3.5k, 7c), or ∼114 (5k, 7d)]. The PEGylated prodrugs 7a-d had a much improved hydrophilicity compared with the non-PEGylated prodrug, Pc-(L-PTX)2. As the PEG length increased, the hydrophilicity of the prodrug increased (log D7.4 values: 1.28, 0.09, -0.24, and -0.59 for 1k, 2k, 3.5k, and 5k PEG prodrugs, respectively). Fluorescence spectral data suggested that the PEGylated prodrugs had good solubility in the culture medium at lower concentrations (<1-2 μM), but showed fluorescence quenching due to limited solubility at higher concentrations (>2 μM). Dynamic light scattering indicated that all of the prodrugs formed nanosized particles in both phosphate-buffered saline and culture medium at a concentration of 5 μM. The PEG length affected both nonspecific and folate receptor (FR)-mediated uptake of the prodrugs. The enhanced cellular uptake was observed for the prodrugs with medium-sized PEGs (1k, 2k, or 3.5k) in FR-positive SKOV-3 cells, but not for the prodrugs with no PEG or with the longest PEG (5k), which suggests the optimal range of PEG length around 1k-3.5k for effective uptake of our prodrug system. Consistent with the cellular uptake pattern, medium-sized PEGylated prodrugs showed more potent phototoxic activity (IC50s, ∼130 nM) than prodrugs with no PEG or the longest PEG (IC50, ∼400 nM). In conclusion, we have developed far-red light-activatable prodrugs with improved water solubility and FR-targeting properties compared with the nontargeted prodrug.
Collapse
Affiliation(s)
- Pritam Thapa
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Mengjie Li
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Radha Karki
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Moses Bio
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Pallavi Rajaputra
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Gregory Nkepang
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Sukyung Woo
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
| | - Youngjae You
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma 73117, United States
- Department
of Chemistry and Biochemistry, University
of Oklahoma, Norman, Oklahoma 73019, United
States
| |
Collapse
|
49
|
Li M, Thapa P, Rajaputra P, Bio M, Peer CJ, Figg WD, You Y, Woo S. Quantitative modeling of the dynamics and intracellular trafficking of far-red light-activatable prodrugs: implications in stimuli-responsive drug delivery system. J Pharmacokinet Pharmacodyn 2017; 44:521-536. [PMID: 28913666 DOI: 10.1007/s10928-017-9543-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022]
Abstract
The combination of photodynamic therapy (PDT) with anti-tumor agents is a complimentary strategy to treat local cancers. We developed a unique photosensitizer (PS)-conjugated paclitaxel (PTX) prodrug in which a PS is excited by near-infrared wavelength light to site-specifically release PTX while generating singlet oxygen (SO) to effectively kill cancer cells with both PTX and SO. The aim of the present study was to identify the determinants influencing the combined efficacy of this light-activatable prodrug, especially the bystander killing effects from released PTX. Using PS-conjugated PTX as a model system, we developed a quantitative mathematical model describing the intracellular trafficking. Dynamics of the prodrug and the model predictions were verified with experimental data using human cancer cells in vitro. The sensitivity analysis suggested that parameters related to extracellular concentration of released PTX, prodrug uptake, target engagement, and target abundance are critical in determining the combined killing efficacy of the prodrug. We found that released PTX cytotoxicity was most sensitive to the retention time of the drug in extracellular space. Modulating drug internalization and conjugating the agents targeted to abundant receptors may provide a new strategy for maximizing the killing capacity of the far-red light-activatable prodrug system. These results provide guidance for the design of the PDT combination study in vivo and have implications for other stimuli-responsive drug delivery systems.
Collapse
Affiliation(s)
- Mengjie Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Pritam Thapa
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Pallavi Rajaputra
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Moses Bio
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Cody J Peer
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - William D Figg
- Clinical Pharmacology Program, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Youngjae You
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
50
|
Zhang FL, Song MR, Yuan GK, Ye HN, Tian Y, Huang MD, Xue JP, Zhang ZH, Liu JY. A Molecular Combination of Zinc(II) Phthalocyanine and Tamoxifen Derivative for Dual Targeting Photodynamic Therapy and Hormone Therapy. J Med Chem 2017; 60:6693-6703. [PMID: 28699738 DOI: 10.1021/acs.jmedchem.7b00682] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The combination of photodynamic therapy and other cancer treatment modalities is a promising strategy to enhance therapeutic efficacy and reduce side effects. In this study, a tamoxifen-zinc(II) phthalocyanine conjugate linked by a triethylene glycol chain has been synthesized and characterized. Having tamoxifen as the targeting moiety, the conjugate shows high specific affinity to MCF-7 breast cancer cells overexpressed estrogen receptors (ERs) and tumor tissues, therefore leading to a cytotoxic effect in the dark due to the cytostatic tamoxifen moiety, and a high photocytotoxicity due to the photosensitizing phthalocyanine unit against the MCF-7 cancer cells. The high photodynamic activity of the conjugate can be attributed to its high cellular uptake and efficiency in generating intracellular reactive oxygen species. Upon addition of exogenous 17β-estradiol as an ER inhibitor, the cellular uptake and photocytotoxicity of the conjugate are reduced significantly. As shown by confocal microscopy, the conjugate is preferentially localized in the lysosomes of the MCF-7 cells.
Collapse
Affiliation(s)
- Feng-Ling Zhang
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China.,College of Pharmaceutical Science, Zhejiang Chinese Medical University , 548 Binwen Road, Hangzhou, 310053, P. R. China
| | - Mei-Ru Song
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Gan-Kun Yuan
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Huan-Nian Ye
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Ye Tian
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Ming-Dong Huang
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Jin-Ping Xue
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| | - Zhi-Hong Zhang
- Fuzhou General Hospital of Nanjing Military Command , 156 West Second Ring Road, Fuzhou 350005, Fujian, P. R. China
| | - Jian-Yong Liu
- State Key Laboratory of Photocatalysis on Energy and Environment & Fujian Engineering Research Center of Functional Materials, College of Chemistry, Fuzhou University , 2 Xueyuan Road, University Town, Fuzhou 350108, Fujian, P. R. China
| |
Collapse
|