1
|
Kolarski D, Szymanski W, Feringa BL. Spatiotemporal Control Over Circadian Rhythms With Light. Med Res Rev 2025; 45:968-984. [PMID: 39757143 PMCID: PMC11976375 DOI: 10.1002/med.22099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
Circadian rhythms are endogenous biological oscillators that synchronize internal physiological processes and behaviors with external environmental changes, sustaining homeostasis and health. Disruption of circadian rhythms leads to numerous diseases, including cardiovascular and metabolic diseases, cancer, diabetes, and neurological disorders. Despite the potential to restore healthy rhythms in the organism, pharmacological chronotherapy lacks spatial and temporal resolution. Addressing this challenge, chrono-photopharmacology, the approach that employs small molecules with light-controlled activity, enables the modulation of circadian rhythms when and where needed. Two approaches-relying on irreversible and reversible drug activation-have been proposed for this purpose. These methodologies are based on photoremovable protecting groups and photoswitches, respectively. Designing photoresponsive bioactive molecules requires meticulous structural optimization to obtain the desired chemical and photophysical properties, and the design principles, detailed guidelines and challenges are summarized here. In this review, we also analyze all the known circadian modulators responsive to light and dissect the rationale following their construction and application to control circadian biology from the protein level to living organisms. Finally, we present the strength of a reversible approach in allowing the modulation of the circadian period and the phase.
Collapse
Affiliation(s)
- Dušan Kolarski
- Max Planck Institute for Multidisciplinary SciencesNanoBioPhotonicsGöttingenGermany
| | - Wiktor Szymanski
- Centre for Systems Chemistry, Stratingh Institute for ChemistryUniversity of GroningenGroningenThe Netherlands
- Department of Radiology, Medical Imaging CenterUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Medicinal Chemistry, Photopharmacology and Imaging, Groningen Research Institute of PharmacyUniversity of GroningenGroningenThe Netherlands
| | - Ben L. Feringa
- Centre for Systems Chemistry, Stratingh Institute for ChemistryUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
2
|
Ojha M, Trivedi P, Banerjee M, Bera M, Dey S, Singh AK, Jana A, Singh NDP. A near infrared light activated phenothiazine based cancer cell specific phototherapeutic system: a synergistic approach to chemo-photothermal therapy. Biomater Sci 2025; 13:1818-1830. [PMID: 39998892 DOI: 10.1039/d4bm01288g] [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: 02/27/2025]
Abstract
In the pursuit of more effective cancer therapies, phototherapy has emerged as a promising approach due to its non-invasive nature and high precision. This study presents the development of a near-infrared (NIR) light-responsive phenothiazine (PTZ) based phototherapeutic system designed for targeted cancer treatment. This phototherapeutic system integrates four crucial elements for enhanced therapeutic efficacy: cancer cell-specific activity, mitochondrial targeting, photothermal conversion, and controlled drug release. The PTZ system utilizes the acidochromic 1,3-oxazine ring, which opens in the acidic tumor microenvironment, forming a positive iminium ion (CN+). This ionic species targets cancer cell mitochondria, ensuring precise localization. Under NIR light irradiation (640 nm), the phototherapeutic system undergoes a red shift in the absorption and reduction in the fluorescence intensity, demonstrating a significant photothermal effect that converts light to heat, thereby inducing tumor cell apoptosis. Furthermore, NIR light triggers the controlled release of the anticancer drug chlorambucil, enabling precise spatiotemporal drug delivery. The closed form of the phototherapeutic system also facilitates drug release upon visible light irradiation (≥410 nm) with high photochemical efficiency. This dual-mode photothermal and photocontrolled drug delivery offers a synergistic approach to cancer therapy, maximizing therapeutic outcomes while minimizing side effects. Our findings underscore the potential of this innovative phototherapeutic system to advance cancer treatment through targeted, controlled, and effective drug delivery.
Collapse
Affiliation(s)
- Mamata Ojha
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| | - Pragya Trivedi
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Moumita Banerjee
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| | - Malabika Bera
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| | - Susmita Dey
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| | - Amit Kumar Singh
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| | - Avijit Jana
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - N D Pradeep Singh
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur 721302, India.
| |
Collapse
|
3
|
Sato K, Okada T, Okada R, Yasui H, Yamada M, Isobe Y, Nishinaga Y, Shimizu M, Koike C, Fukushima R, Takahashi K, Taki S, Kato A, Sato M, Ogura T. Photoinduced Actin Aggregation Involves Cell Death: A Mechanism of Cancer Cell Cytotoxicity after Near-Infrared Photoimmunotherapy. ACS NANO 2025; 19:8338-8356. [PMID: 39964399 PMCID: PMC11887486 DOI: 10.1021/acsnano.5c00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Near-infrared photoimmunotherapy (NIR-PIT) is a cancer treatment modality that uses antibody-photoabsorber (IR700) conjugates to destroy specific cells. The reaction between the antibody and photoabsorber is triggered by NIR-light, and this alters the shape and hydrophilicity of the conjugate. This photochemical reaction is responsible for NIR-PIT-induced cell death; however, the detailed mechanism underlying this effect remains unknown. In this study, we demonstrated that actin filaments underneath the cell membrane play an important role in NIR-PIT-induced cell death and that IR700 mediates the photochemical reaction of the conjugates, leading to actin filament aggregation upon NIR-light irradiation. The destruction of cortical actin beneath the cell plasma membrane allows water to flow into the cell based on osmotic conditions, resulting in cell rupture. This sequence of events may constitute the mechanism of NIR-PIT-induced cell death, making NIR-PIT a promising cancer treatment modality.
Collapse
Affiliation(s)
- Kazuhide Sato
- Nagoya
University Graduate School of Medicine, Nagoya 466-8550, Japan
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- CREST,
JST, Tokyo 102-8666, Japan
- Nagoya
University Institute for Advanced Research, Nagoya 464-8601, Japan
- FOREST-Souhatsu,
JST, Tokyo 102-8666, Japan
| | - Tomoko Okada
- CREST,
JST, Tokyo 102-8666, Japan
- Health and
Medical Research Institute, National Institute of Advanced Industrial
Science and Technology (AIST), Tsukuba 305-8566, Japan
| | - Ryu Okada
- Nagoya
University Graduate School of Medicine, Nagoya 466-8550, Japan
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Hirotoshi Yasui
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Mizuki Yamada
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Yoshitaka Isobe
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Yuko Nishinaga
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Misae Shimizu
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Chiaki Koike
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Rika Fukushima
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Kazuomi Takahashi
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Shunichi Taki
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Ayako Kato
- Nagoya
University Institute for Advanced Research, Advanced Analytical and Diagnostic Imaging Center (AADIC)/Medical
Engineering Unit (MEU), B3 Unit Frontier, Nagoya 466-8550, Japan
| | - Mitsuo Sato
- Division
of Host Defense Sciences, Dept. of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya 461-8673, Japan
| | - Toshihiko Ogura
- CREST,
JST, Tokyo 102-8666, Japan
- Health and
Medical Research Institute, National Institute of Advanced Industrial
Science and Technology (AIST), Tsukuba 305-8566, Japan
| |
Collapse
|
4
|
Janeková H, Fisher S, Šolomek T, Štacko P. Surfing the limits of cyanine photocages one step at a time. Chem Sci 2025; 16:1677-1683. [PMID: 39568875 PMCID: PMC11575602 DOI: 10.1039/d4sc07165d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
Near-infrared light-activated photocages enable controlling molecules with tissue penetrating light. Understanding the structural aspects that govern the photouncaging process is essential to enhancing their efficacy, crucial for practical applications. Here we explore the impact of thermodynamic stabilization on contact ion pairs in cyanine photocages by quaternarization of the carbon reaction centers. This strategy enables the first direct uncaging of carboxylate payloads independent of oxygen, resulting in a remarkable two-orders-of-magnitude enhancement in uncaging efficiency. Our computational analyses reveal that these modifications confer a kinetic instead of thermodynamic effect, reducing ion-ion interactions and allowing complete separation of free ions while inhibiting recombination. We demonstrate that, while thermodynamic stabilization is effective in traditional chromophores operating at shorter wavelengths, it rapidly reaches its thermodynamic limitations in NIR photocages by compromising the photocage stability in the dark. Thanks to these findings, we establish that activation of cyanine photocages is limited to wavelengths of light below 1000 nm. Our work illuminates the path to improving uncaging cross-sections in NIR photocages by prioritizing kinetic trapping and separation of ions.
Collapse
Affiliation(s)
- Hana Janeková
- Department of Chemistry, University of Zurich Winterthurerstrasse 190 CH-8057 Zurich Switzerland
| | - Sergey Fisher
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Tomáš Šolomek
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | - Peter Štacko
- Department of Chemistry, University of Zurich Winterthurerstrasse 190 CH-8057 Zurich Switzerland
| |
Collapse
|
5
|
Zhang H, Xie WC, Yao Y, Tang ZY, Ni WX, Wang B, Gao S, Sessler JL, Zhang JL. Electrostatic Force-Enabled Microneedle Patches that Exploit Photoredox Catalysis for Transdermal Phototherapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:3038-3051. [PMID: 39739671 DOI: 10.1021/acsami.4c18211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Microneedle patches for topical administration of photodynamic therapy (PDT) sensitizers are attractive owing to their safety, selectivity, and noninvasiveness. However, low-efficiency photosensitizer delivery coupled with the limitations of the hypoxic tumor microenvironment remains challenging. To overcome these issues, we developed an effective microneedle patch based on intermolecular electrostatic interactions within a photosensitizer matrix containing a zinc-containing porphyrin analogue, ZnBP (w). This design improved the mechanical strength of the microneedle patch and enhanced the photosensitizer loading efficiency in aqueous environments. A key feature of the system is efficient electron transfer between ZnBP (w) and NADH upon photoirradiation. Electrostatic interactions between ZnBP (w) and NADH were hypothesized to support initial binding and subsequent photoinduced electron transfer, disrupting NADH/NAD+ homeostasis and inducing tumor cell death. The developed microneedle patch demonstrated an antiangiogenesis effect in a vascular malformation model and an antitumor effect in a melanoma mouse model after transdermal administration. This study highlights the benefits of electrostatic interactions in designing microneedle PDT patches and their clinical potential, particularly in reducing systemic phototoxicity.
Collapse
Affiliation(s)
- Hang Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wen-Chuan Xie
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou, Guangdong 515063, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong 515031, China
| | - Yuhang Yao
- College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zi-Yi Tang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Wen-Xiu Ni
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou, Guangdong 515063, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong 515031, China
| | - Bingwu Wang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong 515031, China
| | - Song Gao
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong 515031, China
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712-1224, United States
| | - Jun-Long Zhang
- Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Chemistry and Chemical Engineering Guangdong Laboratory, Shantou, Guangdong 515031, China
| |
Collapse
|
6
|
Wolf I, Storz J, Schultze-Seemann S, Esser PR, Martin SF, Lauw S, Fischer P, Peschers M, Melchinger W, Zeiser R, Gorka O, Groß O, Gratzke C, Brückner R, Wolf P. A new silicon phthalocyanine dye induces pyroptosis in prostate cancer cells during photoimmunotherapy. Bioact Mater 2024; 41:537-552. [PMID: 39246837 PMCID: PMC11378935 DOI: 10.1016/j.bioactmat.2024.07.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/30/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024] Open
Abstract
Photoimmunotherapy (PIT) combines the specificity of antibodies with the cytotoxicity of light activatable photosensitizers (PS) and is a promising new cancer therapy. We designed and synthesized, in a highly convergent manner, the silicon phthalocyanine dye WB692-CB2, which is novel for being the first light-activatable PS that can be directly conjugated via a maleimide linker to cysteines. In the present study we conjugated WB692-CB2 to a humanized antibody with engineered cysteines in the heavy chains that specifically targets the prostate-specific membrane antigen (PSMA). The resulting antibody dye conjugate revealed high affinity and specificity towards PSMA-expressing prostate cancer cells and induced cell death after irradiation with red light. Treated cells exhibited morphological characteristics associated with pyroptosis. Mechanistic studies revealed the generation of reactive oxygen species, triggering a cascade of intracellular events involving lipid peroxidation, caspase-1 activation, gasdermin D cleavage and membrane rupture followed by release of pro-inflammatory cellular contents. In first in vivo experiments, PIT with our antibody dye conjugate led to a significant reduction of tumor growth and enhanced overall survival in mice bearing subcutaneous prostate tumor xenografts. Our study highlights the future potential of the new phthalocyanine dye WB692-CB2 as PS for the fluorescence-based detection and PIT of cancer, including local prostate tumor lesions, and systemic activation of anti-tumor immune responses by the induction of pyroptosis.
Collapse
Affiliation(s)
- Isis Wolf
- Department of Urology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Jonas Storz
- Institute for Organic Chemistry, University of Freiburg, 79104, Freiburg, Germany
| | - Susanne Schultze-Seemann
- Department of Urology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Philipp R Esser
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Allergy Research Group, Department of Dermatology, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Stefan F Martin
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Allergy Research Group, Department of Dermatology, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | - Susan Lauw
- Core Facility Signalling Factory & Robotics, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Peer Fischer
- Max Planck Institute for Medical Research, 69120, Heidelberg, Germany
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, 69120, Heidelberg, Germany
| | - Marie Peschers
- Department of Urology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Wolfgang Melchinger
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Department of Internal Medicine I, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Robert Zeiser
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Department of Internal Medicine I, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Oliver Gorka
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Institute of Neuropathology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Olaf Groß
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Institute of Neuropathology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Reinhard Brückner
- Institute for Organic Chemistry, University of Freiburg, 79104, Freiburg, Germany
| | - Philipp Wolf
- Department of Urology, Medical Center - University of Freiburg, 79106, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| |
Collapse
|
7
|
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
|
8
|
Rigault D, Nizard P, Daniel J, Blanćhard-Desce M, Deprez E, Tauc P, Dhimane H, Dalko PI. Triphenylamine Sensitized 8-Dimethylaminoquinoline: An Efficient Two-Photon Caging Group for Intracellular Delivery. Chemistry 2024; 30:e202401289. [PMID: 38959014 DOI: 10.1002/chem.202401289] [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: 04/01/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024]
Abstract
Triphenylamine-sensitized 8-dimethylaminoquinoline (TAQ) probes showed fair two-photon absorption and fragmentation cross sections in releasing kainate and GABA ligands. The water-soluble PEG and TEG-analogs allowed cell internalization and efficient light-gated liberation of the rhodamine reporter under UV and two-photon (NIR) irradiation conditions.
Collapse
Affiliation(s)
- Delphine Rigault
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Philippe Nizard
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Jonathan Daniel
- Institut des Sciences Moleéculaires, Universite de Bordeaux, Bâtiment A12 351 Cours de la Libération, 33405, TALENCE cedex, France
| | - Mireille Blanćhard-Desce
- Institut des Sciences Moleéculaires, Universite de Bordeaux, Bâtiment A12 351 Cours de la Libération, 33405, TALENCE cedex, France
| | - Eric Deprez
- LBPA, ENS Paris-Saclay, CNRS, Université Paris-Saclay, Gif-sur-Yvette, 91190, France
| | - Patrick Tauc
- LBPA, ENS Paris-Saclay, CNRS, Université Paris-Saclay, Gif-sur-Yvette, 91190, France
| | - Hamid Dhimane
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| | - Peter I Dalko
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, Université Paris Cité, 45 rue des Saints-Pères, 75270, Paris cedex 05, France
| |
Collapse
|
9
|
Capozza M, Digilio G, Gagliardi M, Tei L, Marchesi S, Terreno E, Stefania R. Silicon Phthalocyanines Functionalized with Axial Substituents Targeting PSMA: Synthesis and Preliminary Assessment of Their Potential for PhotoDynamic Therapy of Prostate Cancer. ChemMedChem 2024:e202400218. [PMID: 39082378 DOI: 10.1002/cmdc.202400218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/10/2024] [Indexed: 10/01/2024]
Abstract
Photodynamic therapy (PDT) is a clinical modality based on the irradiation of different diseases, mostly tumours, with light following the selective uptake of a photosensitiser by the pathological tissue. In this study, two new silicon(IV)phtalocyanines (SiPcs) functionalized at both axial positions with a PSMA inhibitor are reported as candidate photosensitizers for PDT of prostate cancer, namely compounds SiPc-PQ(PSMAi)2 and SiPc-OSi(PSMAi)2. These compounds share the same PSMA-binding motif, but differ in the linker that connects the inhibitor moiety to the Si(IV) atom: an alkoxy (Si-O-C) bond for SiPc-PQ(PSMAi)2, and a silyloxy (Si-O-Si) bond for SiPc-OSi(PSMAi)2. Both compounds were synthesized by a facile synthetic route and fully characterized by 2D NMR, mass spectrometry and absorption/fluorescence spectrophotometry. The PDT agents showed a suitable solubility in water, where they essentially exist in monomeric form. SiPc-PQ(PSMAi)2 showed a higher singlet oxygen quantum yield ΦΔ, higher fluorescence quantum yields ΦF and better photostability than SiPc-OSi(PSMAi)2. Both compounds were efficiently taken up by PSMA(+) PC3-PIP cells, but not by PSMA(-) PC3-FLU cells. However, SiPc-PQ(PSMAi)2 showed a more specific photoinduced cytotoxicity in vitro, which is likely attributable to a better stability of its water solutions.
Collapse
Affiliation(s)
- Martina Capozza
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, Torino, 10126, Italy
| | - Giuseppe Digilio
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, Alessandria, 15120, Italy
| | - Michela Gagliardi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, Torino, 10126, Italy
| | - Lorenzo Tei
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, Alessandria, 15120, Italy
| | - Stefano Marchesi
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, Alessandria, 15120, Italy
| | - Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44bis, Torino, 10126, Italy
| | - Rachele Stefania
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, Alessandria, 15120, Italy
| |
Collapse
|
10
|
Cheng Z, Benson S, Mendive-Tapia L, Nestoros E, Lochenie C, Seah D, Chang KY, Feng Y, Vendrell M. Enzyme-Activatable Near-Infrared Hemicyanines as Modular Scaffolds for in vivo Photodynamic Therapy. Angew Chem Int Ed Engl 2024; 63:e202404587. [PMID: 38717316 DOI: 10.1002/anie.202404587] [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: 03/06/2024] [Indexed: 06/21/2024]
Abstract
Photodynamic therapy is an anti-cancer treatment that requires illumination of photosensitizers to induce local cell death. Current near-infrared organic photosensitizers are built from large and non-modular structures that cannot be tuned to improve safety and minimize off-target toxicity. This work describes a novel chemical platform to generate enzyme-activatable near-infrared photosensitizers. We optimized the Se-bridged hemicyanine scaffold to include caging groups and biocompatible moieties, and generated cathepsin-triggered photosensitizers for effective ablation of human glioblastoma cells. Furthermore, we demonstrated that enzyme-activatable Se-bridged hemicyanines are effective photosensitizers for the safe ablation of microtumors in vivo, creating new avenues in the chemical design of targeted anti-cancer photodynamic therapy agents.
Collapse
Affiliation(s)
- Zhiming Cheng
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Sam Benson
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Eleni Nestoros
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Charles Lochenie
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Deborah Seah
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Kai Yee Chang
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| | - Yi Feng
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, The University of Edinburgh, EH4 2XR, Edinburgh, UK
| | - Marc Vendrell
- Centre for Inflammation Research, The University of Edinburgh, EH16 4UU, Edinburgh, UK
- IRR Chemistry Hub, Institute for Regeneration and Repair, The University of Edinburgh, EH16 4UU, Edinburgh, UK
| |
Collapse
|
11
|
Li H, Wang J, Jiao L, Hao E. BODIPY-based photocages: rational design and their biomedical application. Chem Commun (Camb) 2024; 60:5770-5789. [PMID: 38752310 DOI: 10.1039/d4cc01412j] [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: 05/31/2024]
Abstract
Photocages, also known as photoactivated protective groups (PPGs), have been utilized to achieve controlled release of target molecules in a non-invasive and spatiotemporal manner. In the past decade, BODIPY fluorophores, a well-established class of fluorescent dyes, have emerged as a novel type of photoactivated protective group capable of efficiently releasing cargo species upon irradiation. This is due to their exceptional properties, including high molar absorption coefficients, resistance to photochemical and thermal degradation, multiple modification sites, favorable uncaging quantum yields, and highly adjustable spectral properties. Compared to traditional photocages that mainly absorb UV light, BODIPY-based photocages that absorb visible/near-infrared (Vis/NIR) light offer advantages such as deeper tissue penetration and reduced bio-autofluorescence, making them highly suitable for various biomedical applications. Consequently, different types of photoactivated protective groups based on the BODIPY skeleton have been established. This highlight provides a comprehensive overview of the strategies employed to construct BODIPY photocages by substituting leaving groups at different positions within the BODIPY fluorophore, including the meso-methyl position, boron position, 2,6-position, and 3,5-position. Furthermore, the application of these BODIPY photocages in biomedical fields, such as fluorescence imaging and controlled release of active species, is discussed.
Collapse
Affiliation(s)
- Heng Li
- Laboratory of Functional Molecular Solids, Ministry of Education, School of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241002, China.
| | - Jun Wang
- Anhui Engineering Laboratory for Medicinal and Food Homologous Natural Resources Exploration, Department of Chemistry and Pharmaceutical Engineering, Hefei Normal University, Hefei, 230601, China.
| | - Lijuan Jiao
- Laboratory of Functional Molecular Solids, Ministry of Education, School of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241002, China.
| | - Erhong Hao
- Laboratory of Functional Molecular Solids, Ministry of Education, School of Chemistry and Materials Science, Anhui Normal University, Wuhu, 241002, China.
| |
Collapse
|
12
|
Nene LC, Abrahamse H. Phthalocyanine-based probes in alleviating or evading tumour-hypoxia for enhanced photo- and/ sono-mediated therapeutic efficacies. Photodiagnosis Photodyn Ther 2024; 46:104024. [PMID: 38401819 DOI: 10.1016/j.pdpdt.2024.104024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024]
Abstract
This review discusses the possible methods for improving therapeutic efficacies of phthalocyanine (Pcs) -based therapeutic probes in photo- and sono-dynamic therapies under hypoxic conditions. Herein, the structural design strategies including varying the central metal, position substituents and the effects of adjuvant used in supplementing the therapeutics activities of Pcs or formation of NPs are discussed for cancer therapies in hypoxic conditions. Different mechanisms induced for cell death influenced by the compositions of the Pcs-probes are discussed. The focus mainly highlights the oxygen (O2) -dependent mechanisms including methods of supplementing tumour microenvironment O2-concentrations to promote PDT or SDT therapies. Alternatively, O2-independent mechanisms mainly used to evade hypoxia by stimulating anticancer processes that don't require O2 to initiate cell death, such as the Fenton reaction or thermal ablation effects.
Collapse
Affiliation(s)
- Lindokuhle Cindy Nene
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa.
| |
Collapse
|
13
|
Liang X, Qian S, Lou Z, Hu R, Hou Y, Chen PR, Fan X. Near Infrared Light-Triggered Photocatalytic Decaging for Remote-Controlled Spatiotemporal Activation in Living Mice. Angew Chem Int Ed Engl 2023; 62:e202310920. [PMID: 37842955 DOI: 10.1002/anie.202310920] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/30/2023] [Accepted: 10/11/2023] [Indexed: 10/17/2023]
Abstract
Spatiotemporal manipulation of biological processes in living animals using noninvasive, remote-controlled stimuli is a captivating but challenging endeavor. Herein, we present the development of a biocompatible photocatalytic technology termed CAT-NIR, which uses external near infrared light (NIR, 740 nm) to trigger decaging reactions in living mice. The Os(II) terpyridine complex was identified as an efficient NIR photocatalyst for promoting deboronative hydroxylation reactions via superoxide generation in the presence of NIR light, resulting in the deprotection of phenol groups and the release of bioactive molecules under living conditions. The validation of the CAT-NIR system was demonstrated through the NIR-triggered rescue of fluorophores, prodrugs as well as biomolecules ranging from amino acids, peptides to proteins. Furthermore, by combining genetic code expansion and computer-aided screening, CAT-NIR could regulate affibody binding to the cell surface receptor HER2, providing a selective cell tagging technology through external NIR light. In particular, the tissue-penetrating ability of NIR light allowed for facile prodrug activation in living mice, enabling noninvasive, remote-controlled rescue of drug molecules. Given its broad adaptability, this CAT-NIR system may open new opportunities for manipulating the functions of bioactive molecules in living animals using external NIR light with spatiotemporal resolution.
Collapse
Grants
- 22222701, 22077004, 92253301, 21937001, 22137001 National Natural Science Foundation of China
- 22222701, 22077004, 92253301, 22321005, 21937001, 22137001 National Natural Science Foundation of China
- 2019YFA0904201, 2022YFA1304700, 2022YFE0114900 Ministry of Science and Technology
- Z200010, Z221100007422046 Beijing Municipal Science and Technology Commission
- YGLX202338 Beijing Hospitals Authority Clinical Medicine Development Funding
- Li Ge-Zhao Ning Life Science Junior Research Fellowship
Collapse
Affiliation(s)
- Xuan Liang
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Shan Qian
- Department of Pharmaceutical Engineering, College of Food and Bioengineering, Xihua University, Chengdu, 610039, China
| | - Zhizheng Lou
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Renming Hu
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Yuchen Hou
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Peng R Chen
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Xinyuan Fan
- Synthetic and Functional Biomolecules Center, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| |
Collapse
|
14
|
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
|
15
|
Sandberg E, Srambickal CV, Piguet J, Liu H, Widengren J. Local monitoring of photosensitizer transient states provides feedback for enhanced efficiency and targeting selectivity in photodynamic therapy. Sci Rep 2023; 13:16829. [PMID: 37803073 PMCID: PMC10558575 DOI: 10.1038/s41598-023-43625-6] [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: 07/10/2023] [Accepted: 09/26/2023] [Indexed: 10/08/2023] Open
Abstract
Photodynamic therapy (PDT) fundamentally relies on local generation of PDT precursor states in added photosensitizers (PS), particularly triplet and photo-radical states. Monitoring these states in situ can provide important feedback but is difficult in practice. The states are strongly influenced by local oxygenation, pH and redox conditions, often varying significantly at PDT treatment sites. To overcome this problem, we followed local PDT precursor state populations of PS compounds, via their fluorescence intensity response to systematically varied excitation light modulation. Thereby, we could demonstrate local monitoring of PDT precursor states of methylene blue (MB) and IRdye700DX (IR700), and determined their transitions rates under different oxygenation, pH and redox conditions. By fiber-optics, using one fiber for both excitation and fluorescence detection, the triplet and photo-radical state kinetics of locally applied MB and IR700 could then be monitored in a tissue sample. Finally, potassium iodide and ascorbate were added as possible PDT adjuvants, enhancing intersystem crossing and photoreduction, respectively, and their effects on the PDT precursor states of MB and IR700 could be locally monitored. Taken together, the presented procedure overcomes current methodological limitations and can offer feedback, guiding both excitation and PDT adjuvant application, and thereby more efficient and targeted PDT treatments.
Collapse
Affiliation(s)
- Elin Sandberg
- Experimental Biomolecular Physics, Dept. Applied Physics, Royal Institute of Technology (KTH), Albanova Univ Center, 106 91, Stockholm, Sweden
| | - Chinmaya V Srambickal
- Experimental Biomolecular Physics, Dept. Applied Physics, Royal Institute of Technology (KTH), Albanova Univ Center, 106 91, Stockholm, Sweden
| | - Joachim Piguet
- Experimental Biomolecular Physics, Dept. Applied Physics, Royal Institute of Technology (KTH), Albanova Univ Center, 106 91, Stockholm, Sweden
| | - Haichun Liu
- Experimental Biomolecular Physics, Dept. Applied Physics, Royal Institute of Technology (KTH), Albanova Univ Center, 106 91, Stockholm, Sweden
| | - Jerker Widengren
- Experimental Biomolecular Physics, Dept. Applied Physics, Royal Institute of Technology (KTH), Albanova Univ Center, 106 91, Stockholm, Sweden.
| |
Collapse
|
16
|
Rosenberger JE, Xie Y, Fang Y, Lyu X, Trout WS, Dmitrenko O, Fox JM. Ligand-Directed Photocatalysts and Far-Red Light Enable Catalytic Bioorthogonal Uncaging inside Live Cells. J Am Chem Soc 2023; 145:6067-6078. [PMID: 36881718 PMCID: PMC10589873 DOI: 10.1021/jacs.2c10655] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Described are ligand-directed catalysts for live-cell, photocatalytic activation of bioorthogonal chemistry. Catalytic groups are localized via a tethered ligand either to DNA or to tubulin, and red light (660 nm) photocatalysis is used to initiate a cascade of DHTz oxidation, intramolecular Diels-Alder reaction, and elimination to release phenolic compounds. Silarhodamine (SiR) dyes, more conventionally used as biological fluorophores, serve as photocatalysts that have high cytocompatibility and produce minimal singlet oxygen. Commercially available conjugates of Hoechst dye (SiR-H) and docetaxel (SiR-T) are used to localize SiR to the nucleus and microtubules, respectively. Computation was used to assist the design of a new class of redox-activated photocage to release either phenol or n-CA4, a microtubule-destabilizing agent. In model studies, uncaging is complete within 5 min using only 2 μM SiR and 40 μM photocage. In situ spectroscopic studies support a mechanism involving rapid intramolecular Diels-Alder reaction and a rate-determining elimination step. In cellular studies, this uncaging process is successful at low concentrations of both the photocage (25 nM) and the SiR-H dye (500 nM). Uncaging n-CA4 causes microtubule depolymerization and an accompanying reduction in cell area. Control studies demonstrate that SiR-H catalyzes uncaging inside the cell, and not in the extracellular environment. With SiR-T, the same dye serves as a photocatalyst and the fluorescent reporter for microtubule depolymerization, and with confocal microscopy, it was possible to visualize microtubule depolymerization in real time as the result of photocatalytic uncaging in live cells.
Collapse
Affiliation(s)
- Julia E. Rosenberger
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Yixin Xie
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Yinzhi Fang
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Xinyi Lyu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - William S. Trout
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Olga Dmitrenko
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| | - Joseph M. Fox
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19716, USA
| |
Collapse
|
17
|
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
|
18
|
Xiong H, Xu Y, Kim B, Rha H, Zhang B, Li M, Yang GF, Kim JS. Photo-controllable biochemistry: Exploiting the photocages in phototherapeutic window. Chem 2022. [DOI: 10.1016/j.chempr.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
|
19
|
Takahashi K, Yasui H, Taki S, Shimizu M, Koike C, Taki K, Yukawa H, Baba Y, Kobayashi H, Sato K. Near-infrared-induced drug release from antibody-drug double conjugates exerts a cytotoxic photo-bystander effect. Bioeng Transl Med 2022; 7:e10388. [PMID: 36176626 PMCID: PMC9471993 DOI: 10.1002/btm2.10388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 05/21/2022] [Accepted: 07/16/2022] [Indexed: 11/23/2022] Open
Abstract
Ideal cancer treatments specifically target and eradicate tumor cells without affecting healthy cells. Therefore, antibody-based therapies that specifically target cancer antigens can be considered ideal cancer therapies. Antibodies linked with small-molecule drugs (i.e., antibody-drug conjugates [ADCs]) are widely used in clinics as antibody-based therapeutics. However, because tumors express antigens heterogeneously, greater target specificity and stable binding of noncleavable linkers in ADCs limit their antitumor effects. To overcome this problem, strategies, including decreasing the binding strength, conjugating more drugs, and targeting tumor stroma, have been applied, albeit with limited success. Thus, further technological advancements are required to remotely control the ADCs. Here, we described a drug that is photo-releasable from an ADC created via simple double conjugation and its antitumor effects both on target and nontarget tumor cells. Specifically, noncleavable T-DM1 was conjugated with IR700DX to produce T-DM1-IR700. Although T-DM1-IR700 itself is noncleavable, with NIR-light irradiation, it can release DM1-derivatives which elicited antitumor effect in vitro mixed culture and in vivo mixed tumor model which are mimicking heterogeneous tumor-antigen expression same as real clinical tumors. This cytotoxic photo-bystander effect occurred in various types mixed cultures in vitro, and changing antibodies also exerted photo-bystander effects, suggesting that this technology can be used for targeting various specific cancer antigens. These findings can potentially aid the development of strategies to address challenges associated with tumor expression of heterogeneous antigen.
Collapse
Affiliation(s)
- Kazuomi Takahashi
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Hirotoshi Yasui
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Shunichi Taki
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Misae Shimizu
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
| | - Chiaki Koike
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
| | - Kentaro Taki
- Division for Medical Research EngineeringNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
| | - Hiroshi Yukawa
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- Department of Biomolecular EngineeringNagoya University Graduate School of EngineeringNagoyaJapan
| | - Yoshinobu Baba
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- Department of Biomolecular EngineeringNagoya University Graduate School of EngineeringNagoyaJapan
| | - Hisataka Kobayashi
- Molecular Imaging ProgramNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Kazuhide Sato
- Department of Respiratory MedicineNagoya University Graduate School of MedicineShowa‐kuNagoyaJapan
- Nagoya University Institute for Advanced ResearchAdvanced Analytical and Diagnostic Imaging Center (AADIC)/Medical Engineering Unit (MEU), B3 UnitShowa‐kuNagoyaJapan
- Institute of Nano‐Life‐Systems, Institutes of Innovation for Future SocietyNagoya UniversityNagoyaJapan
- FOREST‐Souhatsu, CREST, JSTChiyoda‐kuTokyoJapan
- Nagoya University Institute for Advanced Research, S‐YLCJapan
| |
Collapse
|
20
|
Takakura H, Matsuhiro S, Inanami O, Kobayashi M, Saita K, Yamashita M, Nakajima K, Suzuki M, Miyamoto N, Taketsugu T, Ogawa M. Ligand release from silicon phthalocyanine dyes triggered by X-ray irradiation. Org Biomol Chem 2022; 20:7270-7277. [PMID: 35972402 DOI: 10.1039/d2ob00957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Ligand release from silicon phthalocyanine (SiPc) dyes triggered by near-infrared (NIR) light is a key photochemical reaction involving caged compounds based on SiPc. Although NIR light is relatively permeable compared with visible light, this light can be attenuated by tissue absorption and scattering; therefore, using light to induce photochemical reactions deep inside the body is difficult. Herein, because X-rays are highly permeable and can produce radicals through the radiolysis of water, we investigated whether the axial ligands of SiPcs can be cleaved using X-ray irradiation. SiPcs with different axial ligands (alkoxy, siloxy, oxycarbonyl, and phenoxy groups) were irradiated with X-rays under hypoxic conditions. We found that the axial ligands were cleaved via reactions with hydrated electrons (e-aq), not OH radicals, generated from water in response to X-ray irradiation, and SiPc with alkoxy groups exhibited the highest cleavage efficiency. A quantitative investigation revealed that X-ray-induced axial ligand cleavage proceeds via a radical chain reaction. The reaction is expected to be applicable to the molecular design of X-ray-activatable functional molecules in the future.
Collapse
Affiliation(s)
- Hideo Takakura
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| | - Shino Matsuhiro
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| | - Osamu Inanami
- Graduate School of Veterinary Medicine, Hokkaido University, Kita-ku, Sapporo 060-0818, Japan
| | - Masato Kobayashi
- Department of Chemistry, Faculty of Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan.,WPI-ICReDD, Hokkaido University, Kita-ku, Sapporo 001-0021, Japan
| | - Kenichiro Saita
- Department of Chemistry, Faculty of Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan
| | - Masaki Yamashita
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| | - Kohei Nakajima
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| | - Motofumi Suzuki
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| | - Naoki Miyamoto
- Division of Quantum Science and Engineering, Faculty of Engineering, Hokkaido University, Kita-ku, Sapporo 060-8628, Japan
| | - Tetsuya Taketsugu
- Department of Chemistry, Faculty of Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan.,WPI-ICReDD, Hokkaido University, Kita-ku, Sapporo 001-0021, Japan
| | - Mikako Ogawa
- Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812, Japan.
| |
Collapse
|
21
|
Janeková H, Russo M, Ziegler U, Štacko P. Photouncaging of Carboxylic Acids from Cyanine Dyes with Near-Infrared Light. Angew Chem Int Ed Engl 2022; 61:e202204391. [PMID: 35578980 PMCID: PMC9542589 DOI: 10.1002/anie.202204391] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Indexed: 11/13/2022]
Abstract
Near-infrared light (NIR; 650-900 nm) offers unparalleled advantages as a biocompatible stimulus. The development of photocages that operate in this region represents a fundamental challenge due to the low energy of the excitation light. Herein, we repurpose cyanine dyes into photocages that are available on a multigram scale in three steps and efficiently release carboxylic acids in aqueous media upon irradiation with NIR light up to 820 nm. The photouncaging process is examined using several techniques, providing evidence that it proceeds via photooxidative pathway. We demonstrate the practical utility in live HeLa cells by delivery and release of the carboxylic acid cargo, that was otherwise not uptaken by cells in its free form. In combination with modularity of the cyanine scaffold, the realization of these accessible photocages will fully unleash the potential of the emerging field of NIR-photoactivation and facilitate its widespread adoption outside the photochemistry community.
Collapse
Affiliation(s)
- Hana Janeková
- Department of ChemistryUniversity of ZurichWintherthurerstrasse 1908057ZurichSwitzerland
| | - Marina Russo
- Department of ChemistryUniversity of ZurichWintherthurerstrasse 1908057ZurichSwitzerland
| | - Urs Ziegler
- Center for Microscopy and Image AnalysisUniversity of ZurichWintherthurerstrasse 1908057ZurichSwitzerland
| | - Peter Štacko
- Department of ChemistryUniversity of ZurichWintherthurerstrasse 1908057ZurichSwitzerland
| |
Collapse
|
22
|
Long wavelength-emissive Ru(II) metallacycle-based photosensitizer assisting in vivo bacterial diagnosis and antibacterial treatment. Proc Natl Acad Sci U S A 2022; 119:e2209904119. [PMID: 35914164 PMCID: PMC9371697 DOI: 10.1073/pnas.2209904119] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Ruthenium (Ru) complexes are developed as latent emissive photosensitizers for cancer and pathogen photodiagnosis and therapy. Nevertheless, most existing Ru complexes are limited as photosensitizers in terms of short excitation and emission wavelengths. Herein, we present an emissive Ru(II) metallacycle (herein referred to as 1) that is excited by 808-nm laser and emits at a wavelength of ∼1,000 nm via coordination-driven self-assembly. Metallacycle 1 exhibits good optical penetration (∼7 mm) and satisfactory reactive oxygen species production properties. Furthermore, 1 shows broad-spectrum antibacterial activity (including against drug-resistant Escherichia coli) as well as low cytotoxicity to normal mammalian cells. In vivo studies reveal that 1 is employed in precise, second near-infrared biomedical window fluorescent imaging-guided, photo-triggered treatments in Staphylococcus aureus-infected mice models, with negligible side effects. This work thus broads the applications of supramolecular photosensitizers through the strategy of lengthening their wavelengths.
Collapse
|
23
|
Janeková H, Russo M, Ziegler U, Štacko P. Photouncaging of Carboxylic Acids from Cyanine Dyes with Near‐Infrared Light**. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Hana Janeková
- Department of Chemistry University of Zurich Wintherthurerstrasse 190 8057 Zurich Switzerland
| | - Marina Russo
- Department of Chemistry University of Zurich Wintherthurerstrasse 190 8057 Zurich Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis University of Zurich Wintherthurerstrasse 190 8057 Zurich Switzerland
| | - Peter Štacko
- Department of Chemistry University of Zurich Wintherthurerstrasse 190 8057 Zurich Switzerland
| |
Collapse
|
24
|
Takakura H, Matsuhiro S, Kobayashi M, Goto Y, Harada M, Taketsugu T, Ogawa M. Axial-ligand-cleavable silicon phthalocyanines triggered by near-infrared light toward design of photosensitizers for photoimmunotherapy. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2021.113749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
25
|
Wang S, Zhao J, Zhang L, Zhang C, Qiu Z, Zhao S, Huang Y, Liang H. A Unique Multifunctional Nanoenzyme Tailored for Triggering Tumor Microenvironment Activated NIR-II Photoacoustic Imaging and Chemodynamic/Photothermal Combined Therapy. Adv Healthc Mater 2022; 11:e2102073. [PMID: 34731532 DOI: 10.1002/adhm.202102073] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/27/2021] [Indexed: 12/12/2022]
Abstract
The accurate diagnosis and targeted therapy of malignant tumors face significant challenges. To address these, an oxidized molybdenum polyoxometalate-copper nanocomposite (Ox-POM@Cu) is designed and synthesized here. The doping with Cu determines the formation of oxygen vacancies, which can increase the carrier concentration in Ox-POM@Cu, accelerate electron transfer, and enhance the redox activity, thus playing an efficient catalytic role. The nanocomposite presents unique enzymatic functions characterized by a multielement catalytic activity in the tumor microenvironment (TME). In addition, it can be employed as an NIR-II photoacoustic imaging (PAI) probe and cancer therapy agent. First, it participates in a redox reaction with glutathione (GSH) in tumor tissues, activates the PAI and photothermal therapy functions via NIR-II irradiation, and depletes the GSH supply in cancerous cells. Subsequently, it catalyzes a Fenton-like reaction with H2 O2 in tumor tissues to form hydroxyl radicals, thereby performing a chemodynamic therapy function. The findings show that the developed nanoenzyme is very efficient in the diagnosis and treatment of malignant tumors. This work not only provides a new strategy for the design of TME-induced NIR-II PAI but also presents new insights into enhanced cancer therapy.
Collapse
Affiliation(s)
- Shulong Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Jingjin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Liangliang Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Chaobang Zhang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Zhidong Qiu
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Yong Huang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| | - Hong Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources School of Chemistry and Pharmaceutical Science Guangxi Normal University Guilin 541004 China
| |
Collapse
|
26
|
Xu Y, Tuo W, Yang L, Sun Y, Li C, Chen X, Yang W, Yang G, Stang PJ, Sun Y. Design of a Metallacycle-Based Supramolecular Photosensitizer for In Vivo Image-Guided Photodynamic Inactivation of Bacteria. Angew Chem Int Ed Engl 2022; 61:e202110048. [PMID: 34806264 DOI: 10.1002/anie.202110048] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Indexed: 12/22/2022]
Abstract
Bacterial infection is one of the greatest threats to public health. In vivo real-time monitoring and effective treatment of infected sites through non-invasive techniques, remain a challenge. Herein, we designed a PtII metallacycle-based supramolecular photosensitizer through the host-guest interaction between a pillar[5]arene-modified metallacycle and 1-butyl-4-[4-(diphenylamino)styryl]pyridinium. Leveraging the aggregation-induced emission supramolecular photosensitizer, we improved fluorescence performance and antimicrobial photodynamic inactivation. In vivo studies revealed that it displayed precise fluorescence tracking of S. aureus-infected sites, and in situ performed image-guided efficient PDI of S. aureus without noticeable side effects. These results demonstrated that metallacycle combined with host-guest chemistry could provide a paradigm for the development of powerful photosensitizers for biomedicine.
Collapse
Affiliation(s)
- Yuling Xu
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Wei Tuo
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, UT, 84112, USA
| | - Liang Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yan Sun
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, UT, 84112, USA
| | - Chonglu Li
- Guangxi Key laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoqiang Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing University of Technology, Nanjing, 210009, China
| | - Wenchao Yang
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Guangfu Yang
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| | - Peter J Stang
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, UT, 84112, USA
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology, Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
27
|
Xu Y, Tuo W, Yang L, Sun Y, Li C, Chen X, Yang W, Yang G, Stang PJ, Sun Y. Design of a Metallacycle‐Based Supramolecular Photosensitizer for In Vivo Image‐Guided Photodynamic Inactivation of Bacteria. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202110048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Yuling Xu
- Key Laboratory of Pesticides and Chemical Biology Ministry of Education International Joint Research Center for Intelligent Biosensor Technology and Health College of Chemistry Central China Normal University Wuhan 430079 China
| | - Wei Tuo
- Department of Chemistry University of Utah 315 South 1400 East, Room 2020 Salt Lake City UT 84112 USA
| | - Liang Yang
- Department of Radiology Union Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan 430022 China
| | - Yan Sun
- Department of Chemistry University of Utah 315 South 1400 East, Room 2020 Salt Lake City UT 84112 USA
| | - Chonglu Li
- Guangxi Key laboratory of High-Incidence-Tumor Prevention & Treatment Guangxi Medical University Nanning 530021 China
| | - Xiaoqiang Chen
- State Key Laboratory of Materials-Oriented Chemical Engineering Nanjing University of Technology Nanjing 210009 China
| | - Wenchao Yang
- Key Laboratory of Pesticides and Chemical Biology Ministry of Education International Joint Research Center for Intelligent Biosensor Technology and Health College of Chemistry Central China Normal University Wuhan 430079 China
| | - Guangfu Yang
- Key Laboratory of Pesticides and Chemical Biology Ministry of Education International Joint Research Center for Intelligent Biosensor Technology and Health College of Chemistry Central China Normal University Wuhan 430079 China
| | - Peter J. Stang
- Department of Chemistry University of Utah 315 South 1400 East, Room 2020 Salt Lake City UT 84112 USA
| | - Yao Sun
- Key Laboratory of Pesticides and Chemical Biology Ministry of Education International Joint Research Center for Intelligent Biosensor Technology and Health College of Chemistry Central China Normal University Wuhan 430079 China
| |
Collapse
|
28
|
Farajzadeh N, Güler Kuşçulu N, Yenilmez HY, Bahar D, Altuntas Bayir Z. Anticancer and Biological Properties of New Axially Disubstituted Silicon Phthalocyanines. Dalton Trans 2022; 51:7539-7550. [DOI: 10.1039/d2dt01033j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study reports the synthesis of three novel axially disubstituted silicon phthalocyanines (1-3-Si) and their quaternized phthalocyanines (1-3-QSi). The resulting compounds were characterized by applying spectroscopic techniques including 1H NMR,...
Collapse
|
29
|
Inanami O, Hiraoka W, Goto Y, Takakura H, Ogawa M. EPR Characterisation of Phthalocyanine Radical Anions in Near‐Infrared Photocleavage of the Hydrophilic Axial Ligand of a Photoimmunotherapeutic Reagent, IR700. CHEMPHOTOCHEM 2021. [DOI: 10.1002/cptc.202100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Osamu Inanami
- Faculty of Veterinary Medicine Hokkaido University Sapporo 060-0818 Japan
| | - Wakako Hiraoka
- Department of Physics School of Science and Technology Meiji University Kawasaki 214-8571 Japan
| | - Yuto Goto
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Hideo Takakura
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Mikako Ogawa
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| |
Collapse
|
30
|
Katsurayama Y, Ikabata Y, Maeda H, Segi M, Nakai H, Furuyama T. Direct Near Infrared Light-Activatable Phthalocyanine Catalysts. Chemistry 2021; 28:e202103223. [PMID: 34734432 DOI: 10.1002/chem.202103223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Indexed: 12/29/2022]
Abstract
The high penetration of near-infrared (NIR) light makes it effective for use in selective reactions under light-shielded conditions, such as in sealed reactors and deep tissues. Herein, we report the development of phthalocyanine catalysts directly activated by NIR light to transform small organic molecules. The desired photocatalytic properties were achieved in the phthalocyanines by introducing the appropriate peripheral substituents and central metal. These phthalocyanine photocatalysts promote cross-dehydrogenative-coupling (CDC) under irradiation with 810 nm NIR light. The choice of solvent is important, and a mixture of a reaction-accelerating (pyridine) and -decelerating (methanol) solvents was particularly effective. Moreover, we demonstrate photoreactions under visible-light-shielded conditions through the transmission of NIR light. A combined experimental and computational mechanistic analysis revealed that this NIR reaction does not involve a photoredox-type mechanism with electron transfer, but instead a singlet-oxygen-mediated mechanism with energy transfer.
Collapse
Affiliation(s)
- Yoshino Katsurayama
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Yasuhiro Ikabata
- Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.,Information and Media Center, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan.,Department of Computer Science and Engineering, Toyohashi University of Technology, Toyohashi, Aichi, 441-8580, Japan
| | - Hajime Maeda
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Masahito Segi
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Hiromi Nakai
- Waseda Research Institute for Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.,Department of Chemistry and Biochemistry School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo, 169-8555, Japan.,Elements Strategy Initiative for Catalysts and Batteries (ESICB), Kyoto University Katsura, Kyoto, 615-8520, Japan
| | - Taniyuki Furuyama
- Graduate School of Natural Science and Technology, Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan.,Japan Science and Technology Agency (JST)-PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan
| |
Collapse
|
31
|
Huang X, Chen T, Mu N, Lam HW, Sun C, Yue L, Cheng Q, Gao C, Yuan Z, Wang R. Supramolecular micelles as multifunctional theranostic agents for synergistic photodynamic therapy and hypoxia-activated chemotherapy. Acta Biomater 2021; 131:483-492. [PMID: 34265471 DOI: 10.1016/j.actbio.2021.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
Photodynamic therapy (PDT), where a photosensitizer (under light irradiation) converts molecular oxygen to singlet oxygen to elicit programmed cell death, is a promising cancer treatment modality with a high temporal and spatial resolution. However, only limited cancer treatment efficacy has been achieved in clinical PDT due to the hypoxic conditions of solid tumor microenvironment that limits the generation of singlet oxygen, and PDT process often leads to even more hypoxic microenvironment due to the consumption of oxygens during therapy. Herein, we designed novel supramolecular micelles to co-deliver photosensitizer and hypoxia-responsive prodrug to improve the overall therapeutic efficacy. The supramolecular micelles (CPC) were derived from a polyethylene glycol (PEG) system dually tagged with hydrophilic cucurbit[7]uril (CB[7]) and hydrophobic Chlorin e6 (Ce6), respectively on each end, for synergistic antitumor therapy via PDT of Ce6 and chemotherapy of a hypoxia-responsive prodrug, banoxantrone (AQ4N), loaded into the cavity of CB[7]. In addition, CPC was further modularly functionalized by folate (FA) via strong host-guest interaction between folate-amantadine (FA-ADA) and CB[7] to produce a novel nanoplatform, AQ4N@CPC-FA, for targeted delivery. AQ4N@CPC-FA exhibited enhanced cellular uptake, negligible cytotoxicity and good biocompatibility, and improved intracellular reactive oxygen species (ROS) generation efficiency. More importantly, in vivo evaluation of AQ4N@CPC-FA revealed a synergistic antitumor efficacy between PDT of Ce6 and hypoxia-activated chemotherapy of AQ4N (that can be converted to chemotherapeutic AQ4 for tumor chemotherapy in response to the strengthened hypoxic tumor microenvironment during PDT treatment). This study not only provides a new nanoplatform for synergistic photodynamic-chemotherapeutic treatment, but also offers important new insights to design and development of multifunctional supramolecular drug delivery system. STATEMENT OF SIGNIFICANCE: Photodynamic therapy (PDT) has exhibited a variety of advantages for cancer phototherapy as compared to traditional chemotherapy. However, the unsatisfactory therapeutic efficacy by PDT alone as a result of the enhanced tumor hypoxia during PDT has limited its clinical application. Herein, we designed multifunctional supramolecular micelles to co-deliver photosensitizer and hypoxia-responsive prodrug to improve the overall therapeutic efficacy. The supramolecular micelles are biocompatible and possess strong red absorption, controlled drug release profile, and ultimately enhanced therapeutic outcome via PDT-chemotherapy. This study not only provides a new nanoplatform for synergistic photodynamic-chemotherapeutic treatment of cancer, but also offers important new insights to design and development of multifunctional supramolecular drug delivery tool for multi-modality cancer therapy.
Collapse
Affiliation(s)
- Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China; Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Tunan Chen
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing 400038, China
| | - Ning Mu
- Department of Neurosurgery and Key Laboratory of Neurotrauma, Southwest Hospital, The Third Military Medical University (Army Military Medical University), Chongqing 400038, China
| | - Hou Wang Lam
- Faculty of Life Science and Medicine, King's College London, London, United Kingdom
| | - Chen Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Ludan Yue
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Qian Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Cheng Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China
| | - Zhen Yuan
- Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China.
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, and MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau 999078, China.
| |
Collapse
|
32
|
Ogawa M, Takakura H. Photoimmunotherapy: A new cancer treatment using photochemical reactions. Bioorg Med Chem 2021; 43:116274. [PMID: 34139484 DOI: 10.1016/j.bmc.2021.116274] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/04/2021] [Accepted: 06/05/2021] [Indexed: 12/23/2022]
Abstract
Photoimmunotherapy (PIT) is a new molecular-targeted phototherapy in which administration of an antibody conjugated to IR700 (Ab-IR700, a phthalocyanine derivative) is followed by irradiation with near-infrared light. PIT induces cell death due to cell membrane damage, and the formation of IR700 aggregates on the cell membrane triggered by photochemical reactions is an important mechanism of cell killing. Specifically, water-soluble axial ligands of IR700 are cleaved by the photochemical reaction, and the phthalocyanine stacks up due to the π-π interaction, resulting in the formation of aggregates. In addition, the formation of IR700 radical anions and their protonation are essential for the progress of this photochemical reaction. The elucidation of these mechanisms may lead to the development of more effective compounds in the future. In addition, the optical properties of phthalocyanine are expected to expand the medical application of phthalocyanine derivatives in the future.
Collapse
Affiliation(s)
- Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan.
| | - Hideo Takakura
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| |
Collapse
|
33
|
Liu M, Li C. Recent Advances in Activatable Organic Photosensitizers for Specific Photodynamic Therapy. Chempluschem 2021; 85:948-957. [PMID: 32401421 DOI: 10.1002/cplu.202000203] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/05/2020] [Indexed: 12/18/2022]
Abstract
Photodynamic therapy is an alternative modality for the therapy of diseases such as cancer in a minimally invasive manner. The essential photosensitizer, which acts as a catalyst when absorbing light, converts oxygen into cytotoxic reactive oxygen species that ablate malignant cells through apoptosis and/or necrosis, destroy tumor microvasculature, and stimulate immunity. An activatable photosensitizer whose photoactivity could be turned on by a specific disease biomarker is capable of distinguishing healthy cells from diseased cells, thereby reducing off-target photodamage. In this Minireview, we highlight progress in activatable organic photosensitizers over the past five years, including: (i) biorthogonal activatable BODIPYs; (ii) activatable Se-rhodamine with single-cell resolution; (iii) silicon phthalocyanine targeting oxygen tension; (iv) general D-π-A scaffolds; and (v) AIEgens. The potential challenges and opportunities for developing new types of activatable organic photosensitizers to overcome the hypoxia dilemmas of photodynamic therapy are discussed.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| | - Changhua Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Key Laboratory of Functional Polymer Materials of Ministry of Education, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
34
|
Paraboschi I, Turnock S, Kramer-Marek G, Musleh L, Barisa M, Anderson J, Giuliani S. Near-InfraRed PhotoImmunoTherapy (NIR-PIT) for the local control of solid cancers: Challenges and potentials for human applications. Crit Rev Oncol Hematol 2021; 161:103325. [PMID: 33836238 PMCID: PMC8177002 DOI: 10.1016/j.critrevonc.2021.103325] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/29/2021] [Accepted: 03/26/2021] [Indexed: 01/10/2023] Open
Abstract
Near-InfraRed PhotoImmunoTherapy (NIR-PIT) is a novel cancer-targeted treatment effected by a chemical conjugation between a photosensitiser (e.g. the NIR phthalocyanine dye IRDye700DX) and a cancer-targeting moiety (e.g. a monoclonal antibody, moAb). Delivery of a conjugate in vivo leads to accumulation at the tumour cell surface by binding to cell surface receptors or antigens. Upon deployment of focal NIR-light, irradiation of the conjugate results in a rapid, targeted cell death. However, the mechanisms of action to produce the cytotoxic effects have yet to be fully understood. Herein, we bring together the current knowledge of NIR-PIT from preclinical and clinical studies in a variety of cancers highlighting the key unanswered research questions. Furthermore, we discuss how to enhance the local control of solid cancers using this novel treatment regimen.
Collapse
Affiliation(s)
- Irene Paraboschi
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK
| | - Stephen Turnock
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | | | - Layla Musleh
- Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK; Department of Oncology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, England, UK
| | - Stefano Giuliani
- Wellcome/EPSRC Centre for Interventional & Surgical Sciences, University College London, London, UK; Department of Specialist Neonatal and Pediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
35
|
Zhang Y, Yan C, Zheng Q, Jia Q, Wang Z, Shi P, Guo Z. Harnessing Hypoxia‐Dependent Cyanine Photocages for In Vivo Precision Drug Release. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202017349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yutao Zhang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology and Dynamic Chemistry School of Chemistry and Molecular Engineering East China University of Science & Technology Shanghai 200237 China
| | - Chenxu Yan
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology and Dynamic Chemistry School of Chemistry and Molecular Engineering East China University of Science & Technology Shanghai 200237 China
| | - Qiaoqiao Zheng
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Qian Jia
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education School of Life Science and Technology Xidian University Xi'an Shaanxi 710126 China
| | - Zhongliang Wang
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education School of Life Science and Technology Xidian University Xi'an Shaanxi 710126 China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals Feringa Nobel Prize Scientist Joint Research Center Frontiers Science Center for Materiobiology and Dynamic Chemistry School of Chemistry and Molecular Engineering East China University of Science & Technology Shanghai 200237 China
- State Key Laboratory of Bioreactor Engineering East China University of Science and Technology Shanghai 200237 P. R. China
| |
Collapse
|
36
|
Zhang Y, Yan C, Zheng Q, Jia Q, Wang Z, Shi P, Guo Z. Harnessing Hypoxia-Dependent Cyanine Photocages for In Vivo Precision Drug Release. Angew Chem Int Ed Engl 2021; 60:9553-9561. [PMID: 33569863 DOI: 10.1002/anie.202017349] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/04/2021] [Indexed: 12/13/2022]
Abstract
Photocaging holds promise for the precise manipulation of biological events in space and time. However, current near-infrared (NIR) photocages are oxygen-dependent for their photolysis and lack of timely feedback regulation, which has proven to be the major bottleneck for targeted therapy. Herein, we present a hypoxia-dependent photo-activation mechanism of dialkylamine-substituted cyanine (Cy-NH) accompanied by emissive fragments generation, which was validated with retrosynthesis and spectral analysis. For the first time, we have realized the orthogonal manipulation of this hypoxia-dependent photocaging and dual-modal optical signals in living cells and tumor-bearing mice, making a breakthrough in the direct spatiotemporal control and in vivo feedback regulation. This unique photoactivation mechanism overcomes the limitation of hypoxia, which allows site-specific remote control for targeted therapy, and expands the photo-trigger toolbox for on-demand drug release, especially in a physiological context with dual-mode optical imaging under hypoxia.
Collapse
Affiliation(s)
- Yutao Zhang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Chenxu Yan
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China
| | - Qiaoqiao Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Qian Jia
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Zhongliang Wang
- Engineering Research Center of Molecular-imaging and Neuro-imaging of Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710126, China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Zhiqian Guo
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Science Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai, 200237, China.,State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
37
|
Zhang P, Zeng J, Li Y, Yang C, Meng J, Hou Y, Gao M. Quantitative Mapping of Glutathione within Intracranial Tumors through Interlocked MRI Signals of a Responsive Nanoprobe. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202014348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Peisen Zhang
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
- School of Chemistry and Chemical Engineering Institution University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X) Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions State Key Laboratory of Radiation Medicine and Protection Soochow University Suzhou 215123 China
| | - Yingying Li
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
- School of Chemistry and Chemical Engineering Institution University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Chen Yang
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
- School of Chemistry and Chemical Engineering Institution University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Junli Meng
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
- School of Chemistry and Chemical Engineering Institution University of Chinese Academy of Sciences Beijing 100049 P. R. China
| | - Yi Hou
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
| | - Mingyuan Gao
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics Institute of Chemistry Chinese Academy of Sciences Bei Yi Jie 2, Zhong Guan Cun Beijing 100190 China
- School of Chemistry and Chemical Engineering Institution University of Chinese Academy of Sciences Beijing 100049 P. R. China
- Center for Molecular Imaging and Nuclear Medicine School for Radiological and Interdisciplinary Sciences (RAD-X) Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions State Key Laboratory of Radiation Medicine and Protection Soochow University Suzhou 215123 China
| |
Collapse
|
38
|
Zhang P, Zeng J, Li Y, Yang C, Meng J, Hou Y, Gao M. Quantitative Mapping of Glutathione within Intracranial Tumors through Interlocked MRI Signals of a Responsive Nanoprobe. Angew Chem Int Ed Engl 2021; 60:8130-8138. [PMID: 33283373 DOI: 10.1002/anie.202014348] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Studies reveal that malignant tumors feature uneven distributions of some key biomarkers across the entire tumorous region. Nevertheless, only very limited progress has been made towards non-invasive and quantitative detection of tumor-specific biomarkers in vivo, especially with clinically compatible imaging modalities. Reported here is an Fe3 O4 nanoparticle-based glutathione (GSH) responsive magnetic resonance imaging (MRI) probe that can form particle aggregates within tumors in vivo to give rise to strong GSH concentration dependent interlocked relaxivities. A quantitative correlation between the interlocked MRI signals and local GSH concentration was established, and further applied for mapping the heterogeneous distribution of GSH within an intracranial tumor (2.4 mm × 1.6 mm) in vivo. This methodology will offer a practical route for quantitatively mapping tumor-specific biomarkers in vivo with unlimited detection depth, which largely challenges optical-imaging-based approaches.
Collapse
Affiliation(s)
- Peisen Zhang
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.,School of Chemistry and Chemical Engineering Institution, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Jianfeng Zeng
- Center for Molecular Imaging and Nuclear Medicine, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Yingying Li
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.,School of Chemistry and Chemical Engineering Institution, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Chen Yang
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.,School of Chemistry and Chemical Engineering Institution, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junli Meng
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.,School of Chemistry and Chemical Engineering Institution, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yi Hou
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Mingyuan Gao
- Department Key Laboratory of Colloid Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.,School of Chemistry and Chemical Engineering Institution, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China.,Center for Molecular Imaging and Nuclear Medicine, School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| |
Collapse
|
39
|
Neural and behavioral control in Caenorhabditis elegans by a yellow-light-activatable caged compound. Proc Natl Acad Sci U S A 2021; 118:2009634118. [PMID: 33542099 DOI: 10.1073/pnas.2009634118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Caenorhabditis elegans is used as a model system to understand the neural basis of behavior, but application of caged compounds to manipulate and monitor the neural activity is hampered by the innate photophobic response of the nematode to short-wavelength light or by the low temporal resolution of photocontrol. Here, we develop boron dipyrromethene (BODIPY)-derived caged compounds that release bioactive phenol derivatives upon illumination in the yellow wavelength range. We show that activation of the transient receptor potential vanilloid 1 (TRPV1) cation channel by spatially targeted optical uncaging of the TRPV1 agonist N-vanillylnonanamide at 580 nm modulates neural activity. Further, neuronal activation by illumination-induced uncaging enables optical control of the behavior of freely moving C. elegans without inducing a photophobic response and without crosstalk between uncaging and simultaneous fluorescence monitoring of neural activity.
Collapse
|
40
|
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
|
41
|
Abstract
More than four decades have passed since the first example of a light-activated (caged) compound was described. In the intervening years, a large number of light-responsive derivatives have been reported, several of which have found utility under a variety of in vitro conditions using cells and tissues. Light-triggered bioactivity furnishes spatial and temporal control, and offers the possibility of precision dosing and orthogonal communication with different biomolecules. These inherent attributes of light have been advocated as advantageous for the delivery and/or activation of drugs at diseased sites for a variety of indications. However, the tissue penetrance of light is profoundly wavelength-dependent. Only recently have phototherapeutics that are photoresponsive in the optical window of tissue (600-900 nm) been described. This Review highlights these recent discoveries, along with their limitations and clinical opportunities. In addition, we describe preliminary in vivo studies of prospective phototherapeutics, with an emphasis on the path that remains to be navigated in order to translate light-activated drugs into clinically useful therapeutics. Finally, the unique attributes of phototherapeutics is highlighted by discussing several potential disease applications.
Collapse
|
42
|
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
|
43
|
Wang Y, Zhang C, Wu H, Feng P. Activation and Delivery of Tetrazine-Responsive Bioorthogonal Prodrugs. Molecules 2020; 25:E5640. [PMID: 33266075 PMCID: PMC7731009 DOI: 10.3390/molecules25235640] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/18/2020] [Accepted: 11/26/2020] [Indexed: 02/05/2023] Open
Abstract
Prodrugs, which remain inert until they are activated under appropriate conditions at the target site, have emerged as an attractive alternative to drugs that lack selectivity and show off-target effects. Prodrugs have traditionally been activated by enzymes, pH or other trigger factors associated with the disease. In recent years, bioorthogonal chemistry has allowed the creation of prodrugs that can be chemically activated with spatio-temporal precision. In particular, tetrazine-responsive bioorthogonal reactions can rapidly activate prodrugs with excellent biocompatibility. This review summarized the recent development of tetrazine bioorthogonal cleavage reaction and great promise for prodrug systems.
Collapse
Affiliation(s)
- Yayue Wang
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Chang Zhang
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Haoxing Wu
- Huaxi MR Research Center, Department of Nuclear Medicine, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (C.Z.)
| | - Ping Feng
- Institute of Clinical Trials, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
44
|
Immunomodulatory activity of IR700-labelled affibody targeting HER2. Cell Death Dis 2020; 11:886. [PMID: 33082328 PMCID: PMC7576828 DOI: 10.1038/s41419-020-03077-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/15/2022]
Abstract
There is an urgent need to develop therapeutic approaches that can increase the response rate to immuno-oncology agents. Photoimmunotherapy has recently been shown to generate anti-tumour immunological responses by releasing tumour-associated antigens from ablated tumour cell residues, thereby enhancing antigenicity and adjuvanticity. Here, we investigate the feasibility of a novel HER2-targeted affibody-based conjugate (ZHER2:2395-IR700) selectively to induce cancer cell death in vitro and in vivo. The studies in vitro confirmed the specificity of ZHER2:2395-IR700 binding to HER2-positive cells and its ability to produce reactive oxygen species upon light irradiation. A conjugate concentration- and light irradiation-dependent decrease in cell viability was also demonstrated. Furthermore, light-activated ZHER2:2395-IR700 triggered all hallmarks of immunogenic cell death, as defined by the translocation of calreticulin to the cell surface, and the secretion of ATP, HSP70/90 and HMGB1 from dying cancer cells into the medium. Irradiating a co-culture of immature dendritic cells (DCs) and cancer cells exposed to light-activated ZHER2:2395-IR700 enhanced DC maturation, as indicated by augmented expression of CD86 and HLA-DR. In SKOV-3 xenografts, the ZHER2:2395-IR700-based phototherapy delayed tumour growth and increased median overall survival. Collectively, our results strongly suggest that ZHER2:2395-IR700 is a promising new therapeutic conjugate that has great potential to be applicable for photoimmunotherapy-based regimens.
Collapse
|
45
|
Dcona MM, Mitra K, Hartman MCT. Photocontrolled activation of small molecule cancer therapeutics. RSC Med Chem 2020; 11:982-1002. [PMID: 33479692 PMCID: PMC7513389 DOI: 10.1039/d0md00107d] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 07/19/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer remains one of the leading causes of death worldwide. Conventional treatment of the disease is comprised of chemotherapy, radiation and surgery among other treatment approaches. Chemotherapy is plagued by multiple side-effects caused due to non-specific drug action. Light-based therapies offer an alternative treatment approach that can be fine tuned to achieve the desired effect to treat the disease and address challenges posed by chemotherapeutic side-effects. Photodynamic therapy (PDT) is one of the light mediated treatment modalities that has been successfully applied to treat superficial malignancies with high-efficiency, although its dependence on normoxic conditions limits its efficiency to treat deep-seated tumors. On the other hand, light-sensitive drug-mimetics and drug-release platforms have been deemed efficient in preclinical settings to induce cancer cell death with minimal collateral damage. Drawing from about a decade's worth of examples, we highlight the application of photosensitive molecules as an alternative therapeutic option to PDT and describe their designs that influence the biology of the cancer cells, in turn affecting their viability with high spatio-temporal control.
Collapse
Affiliation(s)
- M Michael Dcona
- Department of Internal Medicine , Virginia Commonwealth University , 1201 East Marshall Street , Richmond , 23298 , Virginia , USA .
- Massey Cancer Center , 401 College St. , Richmond , 23219 , Virginia , USA
| | - Koushambi Mitra
- Massey Cancer Center , 401 College St. , Richmond , 23219 , Virginia , USA
- Department of Chemistry , Virginia Commonwealth University , 1001 W Main St , Richmond , 23284 , Virginia , USA
| | - Matthew C T Hartman
- Massey Cancer Center , 401 College St. , Richmond , 23219 , Virginia , USA
- Department of Chemistry , Virginia Commonwealth University , 1001 W Main St , Richmond , 23284 , Virginia , USA
| |
Collapse
|
46
|
Tang J, Wang L, Loredo A, Cole C, Xiao H. Single-atom replacement as a general approach towards visible-light/near-infrared heavy-atom-free photosensitizers for photodynamic therapy. Chem Sci 2020; 11:6701-6708. [PMID: 32953031 PMCID: PMC7473402 DOI: 10.1039/d0sc02286a] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/30/2020] [Indexed: 12/31/2022] Open
Abstract
Photodynamic therapy has become an emerging strategy for the treatment of cancer. This technology relies on the development of photosensitizers (PSs) that convert molecular oxygen to cytotoxic reactive oxygen species upon exposure to light. In this study, we have developed a facile and general strategy for obtaining visible light/near-infrared-absorbing PSs by performing a simple sulfur-for-oxygen replacement within existing fluorophores. Thionation of carbonyl groups within existing fluorophore cores leads to an improvement of the singlet oxygen quantum yield and molar absorption coefficient at longer wavelengths (deep to 600-800 nm). Additionally, these thio-based PSs lack dark cytotoxicity but exhibit significant phototoxicity against monolayer cancer cells and 3D multicellular tumor spheroids with IC50 in the micromolar range. To achieve tumor-specific delivery, we have conjugated these thio-based PSs to an antibody and demonstrated their tumor-specific therapeutic activity.
Collapse
Affiliation(s)
- Juan Tang
- Department of Chemistry , Rice University , 6100 Main Street , Houston , Texas 77005 , USA .
| | - Lushun Wang
- Department of Chemistry , Rice University , 6100 Main Street , Houston , Texas 77005 , USA .
| | - Axel Loredo
- Department of Chemistry , Rice University , 6100 Main Street , Houston , Texas 77005 , USA .
| | - Carson Cole
- Department of Chemistry , Rice University , 6100 Main Street , Houston , Texas 77005 , USA .
| | - Han Xiao
- Department of Chemistry , Rice University , 6100 Main Street , Houston , Texas 77005 , USA .
- Department of Biosciences , Rice University , 6100 Main Street , Houston , Texas 77005 , USA
- Department of Bioengineering , Rice University , 6100 Main Street , Houston , Texas 77005 , USA
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
Kobayashi M, Harada M, Takakura H, Ando K, Goto Y, Tsuneda T, Ogawa M, Taketsugu T. Theoretical and Experimental Studies on the Near‐Infrared Photoreaction Mechanism of a Silicon Phthalocyanine Photoimmunotherapy Dye: Photoinduced Hydrolysis by Radical Anion Generation. Chempluschem 2020; 85:1959-1963. [DOI: 10.1002/cplu.202000338] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/01/2020] [Indexed: 12/27/2022]
Affiliation(s)
- Masato Kobayashi
- Faculty of Science Hokkaido University Sapporo 060-0810 Japan
- WPI-ICReDD Hokkaido University Sapporo 001-0021 Japan
| | - Mei Harada
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Hideo Takakura
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Kanta Ando
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Yuto Goto
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Takao Tsuneda
- Faculty of Science Hokkaido University Sapporo 060-0810 Japan
- Graduate School of Science Technology, and Innovation Kobe University Kobe 657-8501 Japan
| | - Mikako Ogawa
- Faculty of Pharmaceutical Sciences Hokkaido University Sapporo 060-0812 Japan
| | - Tetsuya Taketsugu
- Faculty of Science Hokkaido University Sapporo 060-0810 Japan
- WPI-ICReDD Hokkaido University Sapporo 001-0021 Japan
| |
Collapse
|
50
|
Vorobev AY, Moskalensky AE. Long-wavelength photoremovable protecting groups: On the way to in vivo application. Comput Struct Biotechnol J 2019; 18:27-34. [PMID: 31890141 PMCID: PMC6920508 DOI: 10.1016/j.csbj.2019.11.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 01/07/2023] Open
Abstract
Photoremovable protective groups (PPGs) and related "caged" compounds have been recognized as a powerful tool in an arsenal of life science methods. The present review is focused on recent advances in design of "caged" compounds which function in red or near-infrared region. The naive comparison of photon energy with that of organic bond leads to the illusion that long-wavelength activation is possible only for weak chemical bonds like N-N. However, there are different means to overcome this threshold and shift the uncaging functionality into red or near-infrared regions for general organic bonds. We overview these strategies, including the novel photochemical and photophysical mechanisms used in newly developed PPGs, singlet-oxygen-mediated photolysis, and two-photon absorption. Recent advances in science places the infrared-sensitive PPGs to the same usability level as traditional ones, facilitating in vivo application of caged compounds.
Collapse
Affiliation(s)
- Aleksey Yu. Vorobev
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, 9 Lavrentiev Ave., Novosibirsk 630090, Russia
- Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russia
| | - Alexander E. Moskalensky
- Novosibirsk State University, Pirogova 2, Novosibirsk 630090, Russia
- Voevodsky Institute of Chemical Kinetics and Combustion SB RAS, Institutskaya str. 3, Novosibirsk 630090, Russia
| |
Collapse
|