1
|
Bel'ko N, Mal'tanova A, Bahdanava A, Lugovski A, Fatykhava S, Shabunya P, Smaliakou A, Poznyak S, Kulahava T, Samtsov M. A near-infrared superoxide generator based on a biocompatible indene-bearing heptamethine cyanine dye. J Mater Chem B 2024; 12:11202-11209. [PMID: 39364565 DOI: 10.1039/d4tb01663g] [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: 10/05/2024]
Abstract
One of the most significant limitations of photodynamic therapy is its reduced efficacy in hypoxic microenvironments, which are typical of the majority of tumors. This work demonstrates that indolenine heptamethine cyanines with different substituents in the polymethine chain and at the terminal heterocycles are effective superoxide generators that can be activated in the near-infrared range. The introduction of an indene moiety into the polymethine chain results in a significant enhancement in photostability compared to dyes with a cyclohexene moiety or an unsubstituted polymethine chain. A hydrophilic indene-bearing heptamethine cyanine dye is shown to be efficiently internalized by Vero E6 cells and to give bright intracellular fluorescence in the 700-850 nm range. Furthermore, the dye generates superoxide anion radicals and induces severe oxidative stress in cells upon activation in the near-infrared range (∼750 nm), ultimately resulting in cell death. The capacity of heptamethine cyanines to generate a superoxide anion radical may prove advantageous for enhancing the efficacy of photodynamic therapy under hypoxic conditions.
Collapse
Affiliation(s)
- Nikita Bel'ko
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| | - Anna Mal'tanova
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya str. 14, Minsk 220006, Belarus
| | - Anastasiya Bahdanava
- Institute for Nuclear Problems, Belarusian State University, Bobruiskaya str. 11, Minsk 220006, Belarus
| | - Anatol Lugovski
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| | - Sviatlana Fatykhava
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Akademika Kuprevicha str. 5-2, Minsk 220141, Belarus
| | - Polina Shabunya
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Akademika Kuprevicha str. 5-2, Minsk 220141, Belarus
| | - Adam Smaliakou
- Department of Physics, Belarusian State University, Bobruiskaya str. 5, Minsk 220006, Belarus
| | - Sergey Poznyak
- Research Institute for Physical Chemical Problems, Belarusian State University, Leningradskaya str. 14, Minsk 220006, Belarus
| | - Tatsiana Kulahava
- Institute for Nuclear Problems, Belarusian State University, Bobruiskaya str. 11, Minsk 220006, Belarus
| | - Michael Samtsov
- A.N. Sevchenko Institute of Applied Physical Problems, Belarusian State University, Kurchatova str. 7, Minsk 220045, Belarus.
| |
Collapse
|
2
|
Bora B, Das N, Bera A, Upadhyay A, Goswami TK. Fluorinated High-Valent Sn(IV) Porphyrins Show Remarkable Photodynamic Activity in Cancer Cells. ChemMedChem 2024; 19:e202400376. [PMID: 39017962 DOI: 10.1002/cmdc.202400376] [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: 05/17/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/18/2024]
Abstract
In recent years, Sn(IV) porphyrins have proven to be excellent choice as photosensitizers for photodynamic therapy. This work reports the synthesis, characterization and photodynamic activity of four high-valent fluorinated Sn(IV) porphyrins having different numbers of F-atoms in the peripheral of meso-phenyl groups viz. (Dichloro)meso-tetrakis(4-fluorophenylporphyrinato)stannic(IV), [Sn(IV)FTPP(Cl)2] or Sn1; (Dichloro)meso-tetrakis(2,4-difluorophenylporphyrinato)stannic(IV), [Sn(IV)2,4-FTPP(Cl)2] or Sn2; (Dichloro)meso-tetrakis(2,6-difluorophenylporphyrinato)stannic(IV), [Sn(IV)2,6-FTPP(Cl)2] or Sn3 and (Dichloro)meso-tetrakis(4-trifluoromethylphenylporphyrinato)stannic(IV), [Sn(IV)CF3TPP(Cl)2] or Sn4. The solid-state structure of Sn1 has been determined by single crystal X-ray diffraction analysis. The increasing number of F-atoms attached to the meso-phenyl positions of the porphyrin framework results in increase of their lipophilicity, singlet oxygen quantum yield (ΦΔ) and photocytotoxicity in A549 (human lung adenocarcinoma cells), MCF-7 and MDA-MB-231 (human breast adenocarcinoma) cells. Sn4 predominantly localize in the mitochondria of A549 cells. The light-induced cell death by the Sn(IV) porphyrins in A549 cells occur primarily via apoptosis.
Collapse
Affiliation(s)
- Bidisha Bora
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| | - Namisha Das
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Tridib K Goswami
- Department of Chemistry, Gauhati University, Guwahati, Assam, 781014, India
| |
Collapse
|
3
|
Dutta J, Bera A, Upadhyay A, Yadav AK, Banerjee S, Sarkar T, Hussain A. Photoactivated Anticancer Activity of Cobalt(III) Complexes with Naturally Occurring Flavonoids Chrysin and Silibinin. Chembiochem 2024; 25:e202400484. [PMID: 38962951 DOI: 10.1002/cbic.202400484] [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: 06/02/2024] [Revised: 06/24/2024] [Accepted: 07/04/2024] [Indexed: 07/05/2024]
Abstract
Photoactive metal complexes of bioessential transition metal ions with natural chelators are gaining interest as photocytotoxic agents for cancer photodynamic therapy (PDT). We report six new cobalt(III) complexes with a mixed-ligand formulation [Co(B)2(L)](ClO4)2 (Co1-Co6), where B represents a N,N-donor α-diimine ligand, namely, phenanthroline (phen; Co1, Co2), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq; Co3, Co4), and dipyrido[3,2-a:2',3'-c]phenazine (dppz; Co5, Co6), and L is the monoanionic form of the naturally occurring flavonoids chrysin (chry; Co1, Co3, Co5) and silibinin (sili; Co2, Co4, Co6). Complexes displayed a d-d absorption band within 500-700 nm and exhibited excellent dark and photostability in solution. Cytotoxicity studies indicated significant activity of Co5 and Co6 against cervical (HeLa) and lung (A549) cancer cells under visible light (400-700 nm) irradiation giving low micromolar IC50 values (2.3-3.4 μM, phototoxicity index~15-30). The complexes demonstrated notably low toxicity against normal HPL1D lung epithelial cells. Flow cytometry assay revealed an apoptotic mode of cell damage triggered by the complexes when irradiated. ROS generation assay indicated the involvement of singlet oxygen species in the cell death mechanism when irradiated with light. Overall, complexes Co5 and Co6 with coordinated dipyridophenazine and flavonoid ligands are potential candidates for cancer PDT applications.
Collapse
Affiliation(s)
- Jyotirmoy Dutta
- Department of Chemistry, Handique Girls' College, Guwahati, Assam, 781001, India
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bengaluru, Karnataka, 560012, India
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bengaluru, Karnataka, 560012, India
| | - Ashish Kumar Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, 221005, India
| | - Tukki Sarkar
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India
| | - Akhtar Hussain
- Department of Chemistry, Handique Girls' College, Guwahati, Assam, 781001, India
| |
Collapse
|
4
|
Holden L, Curley RC, Avella G, Long C, Keyes TE. Targeting Mitochondrial Guanine Quadruplexes for Photoactivatable Chemotherapy in Hypoxic Environments. Angew Chem Int Ed Engl 2024; 63:e202408581. [PMID: 39012206 DOI: 10.1002/anie.202408581] [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: 05/06/2024] [Revised: 06/29/2024] [Accepted: 07/14/2024] [Indexed: 07/17/2024]
Abstract
A first example of a mitochondrial G-quadruplex (mitoG4s) targeted Ru(II) photooxidant complex is reported. The complex, Ru-TAP-PDC3 induces photodamage toward guanine quadruplexes (G4s) located in the mitochondrial genome under hypoxic and normoxic conditions. Ru-TAP-PDC3 shows high affinity for mitoG4s and localises within mitochondria of live HeLa cells. Immunolabelling with anti-G4 antibody, BG4, confirms Ru-TAP-PDC3 associates with G4s within the mitochondria of fixed cells. The complex induces depletion of mtDNA in live cells under irradiation at 405 nm, confirmed by loss of PicoGreen signal from mitochondria. Biochemical studies confirm this process induces apoptosis. The complex shows low dark toxicity and an impressive phototoxicity index (PI) of >89 was determined in Hela under very low intensity irradiation, 5 J/cm2. The phototoxicity is thought to operate through both Type II singlet oxygen and Type III pathways depending on normoxic or hypoxic conditions, from live cell assays and plasmid DNA cleavage. Overall, we demonstrate targeting mitoG4s and mtDNA with a photooxidant is a potent route to achieving apoptosis under hypoxic conditions that can be extended to phototherapy.
Collapse
Affiliation(s)
- Lorcan Holden
- School of Chemical Sciences National Center for Sensor Research, Dublin City University, Dublin, 9, Ireland
| | - Rhianne C Curley
- School of Chemical Sciences National Center for Sensor Research, Dublin City University, Dublin, 9, Ireland
| | - Giuseppe Avella
- School of Chemical Sciences National Center for Sensor Research, Dublin City University, Dublin, 9, Ireland
| | - Conor Long
- School of Chemical Sciences National Center for Sensor Research, Dublin City University, Dublin, 9, Ireland
| | - Tia E Keyes
- School of Chemical Sciences National Center for Sensor Research, Dublin City University, Dublin, 9, Ireland
| |
Collapse
|
5
|
Sawamura R, Masuya-Suzuki A, Iki N. Study on cellular uptake of a hydrophobic near-infrared-absorbing diradical-platinum(II) complex solubilized by albumin using hyperspectral imaging, spectrophotometry, and spectrofluorimetry. ANAL SCI 2024; 40:1857-1865. [PMID: 38896386 PMCID: PMC11422251 DOI: 10.1007/s44211-024-00621-8] [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: 04/07/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Owing to its biopenetrability and minimal invasiveness, near-infrared (NIR) light in the region between 700-1100 nm has attracted attention in cancer diagnosis and therapy. Our group previously reported that the hydrophobic diradical-platinum(II) complex PtL2 is a promising agent for cancer photothermal therapy (L = 3,5-dibromo-1,2-diiminobenzosemiquinonate radical). Because PtL2 does not fluoresce, its intercellular uptake of PtL2 cannot be observed with a fluorescence microscope. In this study, we clarified the uptake and intracellular behavior of PtL2 solubilized by bovine serum albumin (BSA) using hyperspectral imaging enabling spectrophotometric analysis of the image. The spectral changes in the obtained images indicated that the internalization of PtL2 was followed by crystallization of the complex during the long incubation period (> 4 h). Additionally, the binding constant Kb = 5.91 × 104 M-1 could be estimated upon fluorescence quenching analysis of BSA upon binding of PtL2; Kb is two orders of magnitude smaller than that of albumin-common drugs. Considering the small Kb and low solubility of PtL2 in water, we ultimately proposed the internalization path and fate of PtL2 in the cell: release of PtL2 from BSA near cellular membranes and subsequent cellular uptake via membrane permeation followed by saturation, resulting in crystallization.
Collapse
Affiliation(s)
- Ryota Sawamura
- Graduate School of Environmental Studies, Tohoku University, 6-6-07 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8579, Japan.
| | - Atsuko Masuya-Suzuki
- Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8511, Japan
| | - Nobuhiko Iki
- Graduate School of Environmental Studies, Tohoku University, 6-6-07 Aramaki-Aoba, Aoba-Ku, Sendai, 980-8579, Japan.
| |
Collapse
|
6
|
Welsh A, Matshitse R, Khan SF, Nyokong T, Prince S, Smith GS. Trinuclear ruthenium(II) polypyridyl complexes: Evaluation as photosensitizers for enhanced cervical cancer treatment. J Inorg Biochem 2024; 256:112545. [PMID: 38581803 DOI: 10.1016/j.jinorgbio.2024.112545] [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: 12/06/2023] [Revised: 03/23/2024] [Accepted: 03/24/2024] [Indexed: 04/08/2024]
Abstract
Trinuclear ruthenium(II) polypyridyl complexes anchored to benzimidazole-triazine / trisamine scaffolds were investigated as photosensitizers for photodynamic therapy. The trinuclear complexes were noted to produce a significant amount of singlet oxygen in both DMF and aqueous media, are photostable and show appreciable emission quantum yields (ɸem). In our experimental setting, despite the moderate phototoxic activity in the HeLa cervical cancer cell line, the phototoxic indices (PI) of the trinuclear complexes are superior relative to the PIs of a clinically approved photosensitizer, Photofrin®, and the pro-drug 5-aminolevulinic acid (PI: >7 relative to PI: >1 and PI: 4.4 for 5-aminolevulinic acid and Photofrin®, respectively). Furthermore, the ruthenium complexes were noted to show appreciable long-term cytotoxicity upon light irradiation in HeLa cells in a concentration-dependent manner. Consequently, this long-term activity of the ruthenium(II) polypyridyl complexes embodies their ability to reduce the probability of the recurrence of cervical cancer. Taken together, this presents a strong motivation for the development of polymetallic complexes as anticancer agents.
Collapse
Affiliation(s)
- Athi Welsh
- Department of Chemistry, University of Cape Town, Rondebosch 7700, ,South Africa
| | - Refilwe Matshitse
- Institute for Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Saif F Khan
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Faculty of Health Science, Observatory, 7925, South Africa
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Rhodes University, Makhanda 6140, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Faculty of Health Science, Observatory, 7925, South Africa
| | - Gregory S Smith
- Department of Chemistry, University of Cape Town, Rondebosch 7700, ,South Africa.
| |
Collapse
|
7
|
Jana A, Sahoo S, Paul S, Sahoo S, Jayabaskaran C, Chakravarty AR. Photodynamic Therapy with Targeted Release of Boron-Dipyrromethene Dye from Cobalt(III) Prodrugs in Red Light. Inorg Chem 2024; 63:6822-6835. [PMID: 38560761 DOI: 10.1021/acs.inorgchem.4c00244] [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: 04/04/2024]
Abstract
Boron-dipyrromethene (BODIPY) dyes are promising photosensitizers for cellular imaging and photodynamic therapy (PDT) owing to their excellent photophysical properties and the synthetically tunable core. Metalation provides a convenient way to overcome the drawbacks arising from their low aqueous solubility. New photo-/redox-responsive Co(III) prodrug chaperones are developed as anticancer PDT agents for efficient cellular delivery of red-light-active BODIPY dyes. The photobiological activity of heteroleptic Co(III) complexes derived from tris(2-pyridylmethyl)amine (TPA) and acetylacetone-conjugated PEGylated distyryl BODIPY (HL1) or its dibromo analogue (HL2), [CoIII(TPA)(L1/L2)](ClO4)2 (1 and 2), are investigated. The Co(III)/Co(II) redox potential is tuned using the Co(III)-TPA scaffold. Complex 1 displays the in vitro release of BODIPY on red light irradiation. Complex 2, having good singlet oxygen quantum yield (ΦΔ ∼ 0.28 in DMSO), demonstrates submicromolar photocytotoxicity to HeLa cancer cells (IC50 ≈ 0.23 μM) while being less toxic to HPL1D normal cells in red light. Cellular imaging using the emissive complex 1 shows mitochondrial localization and significant penetration into the HeLa tumor spheroids. Complex 2 shows supercoiled DNA photocleavage activity and apoptotic cell death through phototriggered generation of reactive oxygen species. The Co(III)-BODIPY prodrug conjugates exemplify new type of phototherapeutic agents with better efficacy than the organic dyes alone in the phototherapeutic window.
Collapse
Affiliation(s)
- Avishek Jana
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Subhadarsini Sahoo
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Subhadeep Paul
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Chelliah Jayabaskaran
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| |
Collapse
|
8
|
Das N, Bora B, Upadhyay A, Das D, Bera A, Goswami TK. Cu(II) flavonoids as potential photochemotherapeutic agents. Dalton Trans 2024; 53:3316-3329. [PMID: 38260975 DOI: 10.1039/d3dt02663a] [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: 01/24/2024]
Abstract
Flavonoids, naturally derived polyphenolic compounds, have received significant attention due to their remarkable biochemical properties that offer substantial health benefits to humans. In this work, a series of six Cu(II) flavonoid complexes of the formulation [Cu(L1)(L2)](ClO4) where L1 is 3-hydroxy flavone (HF1, 1 and 4), 4-fluoro-3-hydroxy flavone (HF2, 2 and 5), and 2,6-difluoro-3-hydroxy flavone (HF3, 3 and 6); L2 is 1,10-phenanthroline (phen, 1-3) and 2-(anthracen-1-yl)-1H-imidazo[4,5-f][1,10]phenanthroline (aip, 4-6) were successfully synthesized, fully characterized and also evaluated for their in vitro photo-triggered cytotoxicity in cancer cells. The single-crystal X-ray diffraction structure of complex 2 shows square pyramidal geometry around the Cu(II) center. The complexes 1-6 showed quasi-reversible cyclic voltammetric responses for the Cu(II)/Cu(I) couple at ∼-0.230 V with a very large ΔEp value of ∼350-480 mV against the Ag/AgCl reference electrode in DMF-0.1 M tetrabutylammonium perchlorate (TBAP) at a scan rate of 50 mV s-1. The complexes were found to have considerable binding propensity for human serum albumin (HSA) and calf thymus DNA (ct-DNA). The complexes displayed remarkable dose-dependent photocytotoxicity in visible light (400-700 nm) in both A549 (human lung cancer) and MCF-7 (human breast cancer) cell lines while remaining significantly less toxic in darkness. They were found to be much less toxic to HPL1D (immortalized human peripheral lung epithelial) normal cells compared to A549 and MCF-7 cancer cells. Upon exposure to visible light, they generate reactive oxygen species, which are thought to be the main contributors to the death of cancer cells. In the presence of visible light, the complexes predominantly elicit an apoptotic mode of cell death. Complex 6 preferentially localizes in the mitochondria of A549 cells.
Collapse
Affiliation(s)
- Namisha Das
- Department of Chemistry, Gauhati University, Guwahati 781014, Assam, India.
| | - Bidisha Bora
- Department of Chemistry, Gauhati University, Guwahati 781014, Assam, India.
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Dhananjay Das
- Department of Chemistry, Gauhati University, Guwahati 781014, Assam, India.
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Tridib K Goswami
- Department of Chemistry, Gauhati University, Guwahati 781014, Assam, India.
| |
Collapse
|
9
|
Gibbons DJ, Berbiguier Y, Mulvaney JP, Villandier N, Leroy-Lhez S, Williams RM. Free base porphyrin-cyanine dye conjugate: synthesis and optical properties. Photochem Photobiol Sci 2024; 23:163-176. [PMID: 38133701 DOI: 10.1007/s43630-023-00510-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023]
Abstract
The covalent combination of a cyanine dye (IR-783) with a tetraphenyl porphyrin unit through an ether linkage results in a photoactive system capable of producing singlet oxygen. The synthesis, characterization and photophysical properties of the resulting novel free base porphyrin-cyanine conjugate named TPPO-IR-783 (TOI) is reported. Excited state properties were studied in various solvents with differing polarity. The fluorescence is strongly solvent dependent, however this is not the case for singlet oxygen phosphorescence, which is only observed in tetrahydrofuran (THF), when comparing 8 different polar, non-polar and medium-polarity solvents. This novel type of porphyrin-cyanine photosensitizer has the ability to produce singlet oxygen and absorbs light at NIR wavelengths.
Collapse
Affiliation(s)
- Dáire J Gibbons
- LABCiS - UR 22722, Faculty of Sciences and Technology, University of Limoges, 123 Avenue Albert Thomas, 87060, Limoges, France
- Molecular Photonics Group, Van't Hoff Institute for Molecular Sciences (HIMS), Universiteit van Amsterdam, Science Park 904, 1098 XH, Amsterdam, Netherlands
| | - Yann Berbiguier
- LABCiS - UR 22722, Faculty of Sciences and Technology, University of Limoges, 123 Avenue Albert Thomas, 87060, Limoges, France
| | - Jordan P Mulvaney
- Molecular Photonics Group, Van't Hoff Institute for Molecular Sciences (HIMS), Universiteit van Amsterdam, Science Park 904, 1098 XH, Amsterdam, Netherlands
| | - Nicolas Villandier
- LABCiS - UR 22722, Faculty of Sciences and Technology, University of Limoges, 123 Avenue Albert Thomas, 87060, Limoges, France
| | - Stéphanie Leroy-Lhez
- LABCiS - UR 22722, Faculty of Sciences and Technology, University of Limoges, 123 Avenue Albert Thomas, 87060, Limoges, France.
| | - René M Williams
- Molecular Photonics Group, Van't Hoff Institute for Molecular Sciences (HIMS), Universiteit van Amsterdam, Science Park 904, 1098 XH, Amsterdam, Netherlands.
| |
Collapse
|
10
|
Kushwaha R, Singh V, Peters S, Yadav AK, Dolui D, Saha S, Sarkar S, Dutta A, Koch B, Sadhukhan T, Banerjee S. Density Functional Theory-Guided Photo-Triggered Anticancer Activity of Curcumin-Based Zinc(II) Complexes. J Phys Chem B 2023; 127:10266-10278. [PMID: 37988143 DOI: 10.1021/acs.jpcb.3c02382] [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: 11/22/2023]
Abstract
Photodynamic therapy (PDT) has evolved as a new therapeutic modality for cancer treatment with fewer side effects and drug resistance. Curcumin exhibits PDT activity, but its low bioavailability restricts its clinical application. Here, the bioavailability of curcumin was increased by its complex formation with the Zn(II) center. For a structure-activity relationship study, Zn(II)-based complexes (1-3) comprising N^N-based ligands (2,2'-bipyridine in 1 and 2 or 1,10-phenanthroline in 3) and O^O-based ligands (acetylacetone in 1, monoanionic curcumin in 2 and 3) were synthesized and thoroughly characterized. The X-ray structure of the control complex, 1, indicated a square pyramidal shape of the molecules. Photophysical and TD-DFT studies indicated the potential of 2 and 3 as good visible light type-II photosensitizers for PDT. Guided by the TD-DFT studies, the low-energy visible light-triggered singlet oxygen (1O2) generation efficacy of 2 and 3 was explored in solution and in cancer cells. As predicted by the TD-DFT calculations, these complexes produced 1O2 efficiently in the cytosol of MCF-7 cancer cells and ultimately displayed excellent apoptotic anticancer activity in the presence of light. Moreover, the molecular docking investigation showed that complexes 2 and 3 have very good binding affinities with caspase-9 and p-53 proteins and could activate them for cellular apoptosis. Further molecular dynamics simulations confirmed the stability of 3 in the caspase-9 protein binding site.
Collapse
Affiliation(s)
- Rajesh Kushwaha
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Virendra Singh
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Silda Peters
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ashish K Yadav
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Dependu Dolui
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Sukanta Saha
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Sujit Sarkar
- Prescience Insilico Pvt. Ltd., Bengaluru, Karnataka 560066, India
| | - Arnab Dutta
- Department of Chemistry, Indian Institute of Technology Bombay, Powai, Maharashtra 400076, India
| | - Biplob Koch
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Tumpa Sadhukhan
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Samya Banerjee
- Department of Chemistry, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
11
|
Sarkar T, Sahoo S, Neekhra S, Paul M, Biswas S, Babu BN, Srivastava R, Hussain A. A dipyridophenazine Ni(II) dithiolene complex as a dual-acting cancer phototherapy agent activatable within the phototherapeutic window. Eur J Med Chem 2023; 261:115816. [PMID: 37717381 DOI: 10.1016/j.ejmech.2023.115816] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/03/2023] [Accepted: 09/10/2023] [Indexed: 09/19/2023]
Abstract
A combination of photodynamic therapy (PDT) and photothermal therapy (PTT) within the phototherapeutic window (600-900 nm) can lead to significantly enhanced therapeutic outcomes, surpassing the efficacy observed with PDT or PTT alone in cancer phototherapy. Herein, we report a novel small-molecule mixed-ligand Ni(II)-dithiolene complex (Ni-TDD) with a dipyridophenazine ligand, demonstrating potent red-light PDT and significant near-infrared (NIR) light mild-temperature PTT activity against cancer cells and 3D multicellular tumour spheroids (MCTSs). The four-coordinate square planar complex exhibited a moderately intense absorption band (ε ∼ 3700 M-1cm-1) centered around 900 nm and demonstrated excellent dark and photostability in an aqueous phase. Ni-TDD induced a potent red-light (600-720 nm) PDT effect on HeLa cancer cells (IC50 = 1.8 μM, photo irritation factor = 44), triggering apoptotic cell death through efficient singlet oxygen generation. Ni-TDD showed a significant intercalative binding affinity towards double-helical calf thymus DNA, resulting in a binding constant (Kb) ∼ 106 M-1. The complex induced mild hyperthermia and exerted a significant mild-temperature PTT effect on MDA-MB-231 cancer cells upon irradiation with 808 nm NIR light. Simultaneous irradiation of Ni-TDD-treated HeLa MCTSs with red and NIR light led to a remarkable synergistic inhibition of growth, exceeding the effects of individual irradiation, through the generation of singlet oxygen and mild hyperthermia. Ni-TDD displayed minimal toxicity towards non-cancerous HPL1D and L929 cells, even at high micromolar concentrations. This is the first report of a Ni(II) complex demonstrating red-light PDT activity and the first example of a first-row transition metal complex exhibiting combined PDT and PTT effects within the clinically relevant phototherapeutic window. Our findings pave the way for designing and developing metal-dithiolene complexes as dual-acting cancer phototherapy agents using long wavelength light for treating solid tumors.
Collapse
Affiliation(s)
- Tukki Sarkar
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Suditi Neekhra
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India
| | - Milan Paul
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, 500078, Telangana, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, 500078, Telangana, India.
| | - Bathini Nagendra Babu
- Department of Fluoro-Agrochemicals, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500007, India.
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Bombay, Powai, Mumbai, 400076, India.
| | - Akhtar Hussain
- Department of Chemistry, Handique Girls' College, Guwahati, 781001, Assam, India.
| |
Collapse
|
12
|
Yamada T, Komatsu T. Protein-Porphyrin Complex Photosensitizers for Anticancer and Antimicrobial Photodynamic Therapies. ChemMedChem 2023; 18:e202300373. [PMID: 37821798 DOI: 10.1002/cmdc.202300373] [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: 07/18/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/13/2023]
Abstract
Photodynamic therapy (PDT) efficiently induces apoptosis through visible-light irradiation of photosensitizers (PSs) within tumors and microbial cells. Porphyrin analogues serve as widely utilized photosensitizing agents with their therapeutic abilities being governed by molecular structures and central metal ions. However, these macrocyclic compounds tend to agglutinate and form stacks in aqueous environments, resulting in a loss of photochemical activity. To overcome this limitation, encapsulation within liposomes and polymer micelles enables the dispersion of porphyrins as monomolecular entities in aqueous solutions, preventing undesirable deactivation. Recently, the use of reconstituted hemoproteins containing various metal-porphyrins and protein cages incorporating porphyrins has garnered significant interest as a new generation of biocompatible PSs. In this concept paper, we provide a comprehensive review of recent developments and trends of protein-porphyrin complex PSs for applications in anticancer and antimicrobial PDTs.
Collapse
Affiliation(s)
- Taiga Yamada
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Teruyuki Komatsu
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| |
Collapse
|
13
|
Akbar A, Khan S, Chatterjee T, Ghosh M. Unleashing the power of porphyrin photosensitizers: Illuminating breakthroughs in photodynamic therapy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 248:112796. [PMID: 37804542 DOI: 10.1016/j.jphotobiol.2023.112796] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/21/2023] [Accepted: 10/01/2023] [Indexed: 10/09/2023]
Abstract
This comprehensive review provides the current trends and recent developments of porphyrin-based photosensitizers. We discuss their evolution from first-generation to third-generation compounds, including cutting-edge nanoparticle-integrated derivatives, and explores their pivotal role in advancing photodynamic therapy (PDT) for enhanced cancer treatment. Integrating porphyrins with nanoparticles represents a promising avenue, offering improved selectivity, reduced toxicity, and heightened biocompatibility. By elucidating recent breakthroughs, innovative methodologies, and emerging applications, this review provides a panoramic snapshot of the dynamic field, addressing challenges and charting prospects. With a focus on harnessing reactive oxygen species (ROS) through light activation, PDT serves as a minimally invasive therapeutic approach. This article offers a valuable resource for researchers, clinicians, and PDT enthusiasts, highlighting the potential of porphyrin photosensitizers to improve the future of cancer therapy.
Collapse
Affiliation(s)
- Alibasha Akbar
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Syamantak Khan
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
| | - Tanmay Chatterjee
- Department of Chemistry, Birla Institute of Technology & Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Hyderabad 500078, Telangana, India
| | - Mihir Ghosh
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
14
|
Curley R, Burke CS, Gkika KS, Noorani S, Walsh N, Keyes TE. Phototoxicity of Tridentate Ru(II) Polypyridyl Complex with Expanded Bite Angles toward Mammalian Cells and Multicellular Tumor Spheroids. Inorg Chem 2023; 62:13089-13102. [PMID: 37535942 PMCID: PMC10428208 DOI: 10.1021/acs.inorgchem.3c01982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 08/05/2023]
Abstract
Tridentate ligand-coordinated ruthenium (II) polypyridyl complexes with large N-Ru-N bite angles have been shown to promote ligand field splitting and reduce singlet-triplet state mixing leading to dramatically extended emission quantum yields and lifetimes under ambient conditions. These effects are anticipated to enhance their photoinduced singlet oxygen production, promoting prospects for such complexes as type II phototherapeutics. In this contribution, we examined this putative effect for [Ru(bqp)(bqpCOOEt)]2+, Ru-bqp-ester, a heteroleptic complex containing bqp = [2,6-bi(quinolin-8-yl)pyridine], a well-established large bite angle tridentate ligand, as well as its peptide conjugates [Ru(bqp)(bqpCONH-ahx-FrFKFrFK(Ac)-CONH2)]5+ (Ru-bqp-MPP) and [Ru(bqp) (bqp)(CONH-ahx-RRRRRRRR-CONH2)]10+ (Ru-bqp-R8) that were prepared in an effort to promote live cell/tissue permeability and targeting of the parent. Membrane permeability of both parent and peptide conjugates were compared across 2D cell monolayers; A549, Chinese hamster ovary, human pancreatic cancer (HPAC), and 3D HPAC multicellular tumor spheroids (MCTS) using confocal microscopy. Both the parent complex and peptide conjugates showed exceptional permeability with rapid uptake in both 2D and 3D cell models but with little distinction in permeability or distribution in cells between the parent or peptide conjugates. Unexpectedly, the uptake was temperature independent and so attributed to passive permeation. Both dark and photo-toxicity of the Ru(II) complexes were assessed across cell types, and the parent showed notably low dark toxicity. In contrast, the parent and conjugates were found to be highly phototoxic, with impressive phototoxic indices (PIs) toward HPAC cell monolayers in particular, with PI values ranging from ∼580 to 760. Overall, our data indicate that the Ru(II) parent complex and its peptide conjugates show promise at both cell monolayers and 3D MCTS as photosensitizers for photodynamic therapy.
Collapse
Affiliation(s)
- Rhianne
C. Curley
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Christopher S. Burke
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Karmel S. Gkika
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Sara Noorani
- National
Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Naomi Walsh
- National
Institute for Cellular Biotechnology, School of Biotechnology, Dublin City University, Dublin 9 D09 NA55, Ireland
| | - Tia E. Keyes
- School
of Chemical Sciences and National Centre for Sensor Research, Dublin City University, Dublin 9 D09 NA55, Ireland
| |
Collapse
|
15
|
Lima E, Reis LV. Photodynamic Therapy: From the Basics to the Current Progress of N-Heterocyclic-Bearing Dyes as Effective Photosensitizers. Molecules 2023; 28:5092. [PMID: 37446758 DOI: 10.3390/molecules28135092] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/16/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Photodynamic therapy, an alternative that has gained weight and popularity compared to current conventional therapies in the treatment of cancer, is a minimally invasive therapeutic strategy that generally results from the simultaneous action of three factors: a molecule with high sensitivity to light, the photosensitizer, molecular oxygen in the triplet state, and light energy. There is much to be said about each of these three elements; however, the efficacy of the photosensitizer is the most determining factor for the success of this therapeutic modality. Porphyrins, chlorins, phthalocyanines, boron-dipyrromethenes, and cyanines are some of the N-heterocycle-bearing dyes' classes with high biological promise. In this review, a concise approach is taken to these and other families of potential photosensitizers and the molecular modifications that have recently appeared in the literature within the scope of their photodynamic application, as well as how these compounds and their formulations may eventually overcome the deficiencies of the molecules currently clinically used and revolutionize the therapies to eradicate or delay the growth of tumor cells.
Collapse
Affiliation(s)
- Eurico Lima
- CQ-VR-Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal
| | - Lucinda V Reis
- CQ-VR-Chemistry Centre of Vila Real, University of Trás-os-Montes and Alto Douro, Quinta de Prados, 5001-801 Vila Real, Portugal
| |
Collapse
|
16
|
Skoczynska A, Lewinski A, Pokora M, Paneth P, Budzisz E. An Overview of the Potential Medicinal and Pharmaceutical Properties of Ru(II)/(III) Complexes. Int J Mol Sci 2023; 24:ijms24119512. [PMID: 37298471 DOI: 10.3390/ijms24119512] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
This review examines the existing knowledge about Ru(II)/(III) ion complexes with a potential application in medicine or pharmacy, which may offer greater potential in cancer chemotherapy than Pt(II) complexes, which are known to cause many side effects. Hence, much attention has been paid to research on cancer cell lines and clinical trials have been undertaken on ruthenium complexes. In addition to their antitumor activity, ruthenium complexes are under evaluation for other diseases, such as type 2 diabetes, Alzheimer's disease and HIV. Attempts are also being made to evaluate ruthenium complexes as potential photosensitizers with polypyridine ligands for use in cancer chemotherapy. The review also briefly examines theoretical approaches to studying the interactions of Ru(II)/Ru(III) complexes with biological receptors, which can facilitate the rational design of ruthenium-based drugs.
Collapse
Affiliation(s)
- Anna Skoczynska
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Andrzej Lewinski
- Department of Endocrinology and Metabolic Diseases, Medical University of Lodz, 93-338 Lodz, Poland
| | - Mateusz Pokora
- International Center of Research on Innovative Biobased Materials (ICRI-BioM)-International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Piotr Paneth
- International Center of Research on Innovative Biobased Materials (ICRI-BioM)-International Research Agenda, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
- Institute of Applied Radiation Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924 Lodz, Poland
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Medical University of Lodz, 90-151 Lodz, Poland
| |
Collapse
|
17
|
Sahoo S, Pathak S, Kumar A, Nandi D, Chakravarty AR. Lysosome directed red light photodynamic therapy using glycosylated iron-(III) conjugates of boron-dipyrromethene. J Inorg Biochem 2023; 244:112226. [PMID: 37105008 DOI: 10.1016/j.jinorgbio.2023.112226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023]
Abstract
To overcome the drawbacks associated with chemotherapeutic and porphyrin-based photodynamic therapy (PDT) agents, the use of BODIPY (boron-dipyrromethene) scaffold has gained prominence in designing a new generation of photosensitizers-cum-cellular imaging agents. However, their poor cell permeability and limited solubility in aqueous medium inhibits the in-vitro application of their organic form. This necessitates the development of metal-BODIPY conjugates with improved physiological stability and enhanced therapeutic efficacy. We have designed two iron(III)-BODIPY conjugates, [Fe(L1/2)(L3)Cl] derived from benzyl-dipicolylamine and its glycosylated analogue along with a BODIPY-tagged catecholate. The complexes showed intense absorption bands (ε ∼ 55,000 M-1 cm-1) and demonstrated apoptotic PDT activity upon red-light irradiation (30 J/cm2, 600-720 nm). The complex with singlet oxygen quantum yield value of ∼0.34 gave sub-micromolar IC50 (half-maximal inhibitory concentration) value (∼0.08 μM) in both HeLa and H1299 cancer cells with a photocytotoxicity index value of >1200. Both the complexes were found to have significantly lower cytotoxic effects in non-cancerous HPL1D (human peripheral lung epithelial) cells. Singlet oxygen was determined to be the prime reactive oxygen species (ROS) responsible for cell damage from pUC19 DNA photo-cleavage studies, 1,3-diphenylisobenzofuran and SOSG (Singlet Oxygen Sensor Green) assays. Cellular imaging studies showed excellent fluorescence from complex 2 within 4 h, with localization in lysosomes. Significant drug accumulation into the core of 3D multicellular tumor spheroids was observed within 8 h from intense in-vitro emission. The complexes exemplify iron-based targeted PDT agents and show promising results as potential transition metal-based drugs for ROS mediated red light photocytotoxicity with low dosage requirement.
Collapse
Affiliation(s)
- Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Sanmoy Pathak
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Arun Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India.
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore 560012, India.
| |
Collapse
|
18
|
Yamada T, Morita Y, Takada R, Funamoto M, Okamoto W, Kohno M, Komatsu T. Zinc Substituted Myoglobin-Albumin Fusion Protein: A Photosensitizer for Cancer Therapy. Chemistry 2023; 29:e202203952. [PMID: 36689636 DOI: 10.1002/chem.202203952] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/18/2023] [Accepted: 01/23/2023] [Indexed: 01/25/2023]
Abstract
Myoglobin combined with human serum albumin (Mb-HSA) can be produced using yeast Pichia pastoris as a host strain, with secretion into the culture medium. This Mb-HSA fusion protein possesses identical O2 binding affinity to that of naked Mb. The Mb unit is reconstituted with a zinc(II) protoporphyrin IX, yielding (zinc substituted Mb)-HSA, ZnMb-HSA. The photophysical property and singlet O2 generation ability of ZnMb-HSA are equivalent to those of ZnMb. In vitro cell experiments revealed that ZnMb-HSA acts as a superior photosensitizer for photodynamic cancer therapy. It is noteworthy that ZnMb-HSA shows long circulation lifetime in vivo.
Collapse
Affiliation(s)
- Taiga Yamada
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Yoshitsugu Morita
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Ryoya Takada
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Mizuki Funamoto
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Wataru Okamoto
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| | - Mitsutomo Kohno
- Department of General Thoracic Surgery, Saitama Medical Center, Saitama Medical University, 1981 Kamoda, Kawagoe-shi, Saitama, 350-8550, Japan
| | - Teruyuki Komatsu
- Department of Applied Chemistry, Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo, 112-8551, Japan
| |
Collapse
|
19
|
Bera A, Gautam S, Sahoo S, Pal AK, Kondaiah P, Chakravarty AR. Red light active Pt(iv)-BODIPY prodrug as a mitochondria and endoplasmic reticulum targeted chemo-PDT agent. RSC Med Chem 2022; 13:1526-1539. [PMID: 36561074 PMCID: PMC9749958 DOI: 10.1039/d2md00225f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/22/2022] [Indexed: 12/25/2022] Open
Abstract
A cisplatin-based platinum(iv) prodrug, [Pt(NH3)2Cl2(OH)(L 1 )], having L 1 as a red-light active boron-dipyrromethene (BODIPY) pendant, was synthesized and characterized and its application as a chemo-cum-photodynamic therapy agent was studied. Me-L 1 as the ligand precursor is structurally characterized. The complex displayed an intense absorption band near 650 nm (ε ∼ 8.8 × 104 dm3 mol-1 cm-1) in 1 : 1 (v/v) DMSO/DPBS. It showed an emission band at 674 nm (λ ex = 630 nm) with a fluorescence quantum yield (Φ F) value of 0.37. In red light (600-720 nm), it generated singlet oxygen as evidenced from the 1,3-diphenylisobenzofuran (DPBF) titration experiment giving a singlet oxygen quantum yield (Φ Δ) value of 0.28 in DMSO. The mechanistic pUC19 DNA photocleavage study and singlet oxygen sensor green (SOSG) assay ascertained its ability to generate singlet oxygen in both extracellular and intracellular media by a type-II photo-process. The complex exhibited high stability in the dark, but on red-light irradiation, it displayed rapid activation in the presence of a reducing environment. It displayed remarkable apoptotic photocytotoxicity with half-maximal inhibitory concentration (IC50) ranging from 0.58 to 0.76 μM in human cervical cancer (HeLa) and breast cancer (MCF-7) cells with a respective photo-cytotoxicity index value of >172 and >131. The photodynamic activity was significantly less in non-cancerous human peripheral lung epithelial (HPL1D) cells. The emissive complex showed localization in the mitochondria and endoplasmic reticulum (ER) with a similar Pearson's correlation coefficient value, making it a dual organelle-targeted therapeutic agent. JC-1, fluo-4-AM and annexin V-FITC/propidium iodide assays in HeLa cells showed cellular apoptosis by arresting cells in the sub-G1 phase via mitochondrial dysfunction and ER stress.
Collapse
Affiliation(s)
- Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Srishti Gautam
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560012 India +91 80 22932688
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Apurba Kumar Pal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science Bangalore 560012 India +91 80 22932688
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India +91 80 22932533
| |
Collapse
|
20
|
Banaspati A, Ramu V, Raza MK, Goswami TK. Copper(ii) curcumin complexes for endoplasmic reticulum targeted photocytotoxicity. RSC Adv 2022; 12:30722-30733. [PMID: 36349155 PMCID: PMC9606729 DOI: 10.1039/d2ra04813b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/01/2022] [Indexed: 08/10/2023] Open
Abstract
Three copper(ii) complexes viz. [Cu(cur)(L)(ClO4)] (1-3), where Hcur is curcumin and L is 1,10-phenanthroline (phen, 1), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq, 2), or dipyrido[3,2-a:2',3'-c]phenazine (dppz, 3) were synthesized, fully characterized by various physicochemical methods and evaluated for their light-assisted chemotherapeutic potential. The complexes [Cu(acac)(L)(ClO4)] (4-6), where Hacac is acetylacetone and L is phen (in 4), dpq (in 5) and dppz (in 6), were synthesized and used as controls. The solid state structures of complexes 4 and 5 were determined by single crystal X-ray diffraction. The curcumin complexes (1-3) were redox inactive at the copper centre, whereas the acetylacetonato complexes (4-6) displayed a Cu(ii)/Cu(i) couple at ∼0.1 V vs. Ag/AgCl reference electrode in DMF. Complexes 1-3 showed an intense curcumin-based band at ∼440 nm in DMF-Tris-HCl buffer (pH = 7.2) (1 : 9 v/v) which masks the copper based d-d band. The complexes bind to human serum albumin (HSA) with moderate efficacy. They also displayed significant binding affinity for calf-thymus (CT) DNA. The lipophilic curcumin complexes show remarkable visible light induced cytotoxicity (IC50 = ∼4 μM) with high phototoxic indices (PI) with low dark toxicity in human cervical carcinoma (HeLa) and human lung carcinoma (A549) cells. The corresponding acetylacetonato controls (4-6) did not show significant cytotoxicity in the dark or light. DCFDA and annexin V-FITC/PI assays using flow cytometry confirm the induction of significant apoptosis in cancer cells via generation of cytotoxic reactive oxygen species upon photoactivation. Confocal microscopic images using complex 3 demonstrate localization of the complexes predominantly in the endoplasmic reticulum of HeLa cells.
Collapse
Affiliation(s)
- Atrayee Banaspati
- Department of Chemistry, Gauhati University Guwahati 781014 Assam India
| | - Vanitha Ramu
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore 560012 India
| | - Tridib K Goswami
- Department of Chemistry, Gauhati University Guwahati 781014 Assam India
| |
Collapse
|
21
|
Luo S, Luo X, Wang X, Li L, Liu H, Mo B, Gan H, Sun W, Wang L, Liang H, Yu S. Tailoring Multifunctional Small Molecular Photosensitizers to In Vivo Self-Assemble with Albumin to Boost Tumor-Preferential Accumulation, NIR Imaging, and Photodynamic/Photothermal/Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201298. [PMID: 35652504 DOI: 10.1002/smll.202201298] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/30/2022] [Indexed: 06/15/2023]
Abstract
Cancer immunotherapy has great potential in tumor eradication and metastasis suppression. However, systemic administration of immune adjuvants and inadequate specificity in cancer treatment, lead to restricted therapeutic benefits and potential immune-related side effects in clinical settings. In this report, the synthesis of various lengths of heptamethine cyanine small molecules to act as multifunctional photosensitizers (PS) for tumor-specific accumulation, near-infrared (NIR) fluorescent imaging, and photodynamic/photothermal/immunotherapy is optimized. In particular, it is demonstrated that C8, which contains eight carbons on two N-alkyl side chains, efficiently self-assembles with albumin to form nanosized dye-albumin complexes. This feature facilitates C8 in vivo self-assembly to remarkably improve its water-solubility, NIR fluorescent emission, long-term blood circulation, as well as tumor-specific accumulation. More importantly, C8 not only exhibits a superior phototherapeutic effect on primary tumors, but also elicits secretion of damage associated molecular patterns, cytokine secretion, dendritic cell maturation, and cytotoxic T lymphocytes activation, ultimately triggering a sufficient antitumor immune response to suppress growths of distant and metastatic tumors. Hence, this multifunctional small molecular PS is characterized with excellent tumor-preferential accumulation, imaging-guided laser irradiation, and phototherapy-induced in situ antitumor immune response, providing a prospective future of its use in tumor-targeting immunotherapy.
Collapse
Affiliation(s)
- Shenglin Luo
- Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xi Luo
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Xiaojiao Wang
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Lian Li
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Huiguo Liu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Banghui Mo
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Hongbo Gan
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Chongqing Key Laboratory of Cytomics, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Liting Wang
- Biomedical Analysis Center, Chongqing Key Laboratory of Cytomics, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Houjie Liang
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| | - Songtao Yu
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), 30 Gaotanyan Street, Chongqing, 400038, China
| |
Collapse
|
22
|
Paul S, Pathak S, Sahoo S, Maji RC, Bhattacharyya U, Nandi D, Chakravarty AR. Bichromophoric ruthenium(II) bis-terpyridine-BODIPY based photosensitizers for cellular imaging and photodynamic therapy. Dalton Trans 2022; 51:10392-10405. [PMID: 35758169 DOI: 10.1039/d2dt01137a] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Two multichromophoric homoleptic ruthenium(II) complexes [Ru(tpy-BODIPY)2]Cl2 (complexes 1 and 2, tpy = 4-phenyl-2,2:6,2-terpyridine, BODIPY = boron-dipyrromethene) were prepared, characterized and their phototherapeutic activity and bioimaging properties were studied. The complexes having structural similarity differ only by a phenylethynyl linker, and its overall influence on their physicochemical and photobiological behavior was evaluated. The terpyridine-BODIPY ligand L1 was structurally characterized by X-ray crystallography. The complexes showed intense absorption near 500 nm (ε: ∼1.5 × 105 M-1 cm-1 in DMSO), have a high singlet oxygen quantum yield (ΦΔ: ∼0.6 in DMSO), and displayed low photobleaching thus making them suitable for PDT applications. The complexes showed high DNA binding affinity and induced DNA damage on light activation via multiple types of ROS production. Confocal laser scanning microscopy experiments revealed their incorporation in the cancer cells and complex 1 predominantly accumulated in lysosomes. The complexes displayed a significant PDT effect in cancerous cells with visible light activation with a high photocytotoxicity index (PI) value in HeLa cells. Both type-I and type-II photosensitization processes were involved in the PDT effect. The photodynamic action of complex 2 initiated cellular apoptosis. Finally, their diagnostic potential was evaluated against clinically relevant 3D multicellular tumor spheroids (MCTs).
Collapse
Affiliation(s)
- Subhadeep Paul
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Sanmoy Pathak
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Ram Chandra Maji
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Utso Bhattacharyya
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
23
|
Durán-Hernández J, Muñoz-Rugeles L, Guzmán-Méndez Ó, M Reza M, Cadena-Caicedo A, García-Montalvo V, Peón J. Sensitization of Nd 3+ Luminescence by Simultaneous Two-Photon Excitation through a Coordinating Polymethinic Antenna. J Phys Chem A 2022; 126:2498-2510. [PMID: 35436116 DOI: 10.1021/acs.jpca.2c01052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We have designed and synthesized two new cyaninic Nd3+ complexes where the lanthanide emission can be induced from simultaneous two-photon absorption followed by energy migration. These complexes correspond to a molecular design that uses an antenna ligand formed by the functionalization of a heptamethine dye with 5-ol-phenanthroline or 4-phenyl-terpyridine derivatives. These complexes employ the important nonlinear optical properties of symmetric polymethines to sensitize the lanthanide ion. We verified that simultaneous biphotonic excitation indirectly induces the 4F3/2 → 4I11/2 Nd3+ emission using femtosecond laser pulses tuned below the first electronic transition of the antenna. The simultaneous two-photon excitation events initially form the nonlinear-active second excited singlet of the polymethine antenna, which rapidly evolves into its first excited singlet. This state in turn induces the formation of the emissive Nd3+ states through energy transfer. The role of the first excited singlet of the antenna as the donor state in this process was verified through time resolution of the antenna's fluorescence. These measurements also provided the rates for antenna-lanthanide energy transfer, which indicate that the phenanthroline-type ligand is approximately five times more efficient for energy transfer than the phenyl-terpyridine derivative due to their relative donor-acceptor distances. The simultaneous two-photon excitation of this polymethine antenna allows for high spatial localization of the Nd3+excitation events.
Collapse
Affiliation(s)
- Jesús Durán-Hernández
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Leonardo Muñoz-Rugeles
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Óscar Guzmán-Méndez
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Mariana M Reza
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Andrea Cadena-Caicedo
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | | | - Jorge Peón
- Instituto de Química, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| |
Collapse
|
24
|
Bera A, Gautam S, Raza MK, Pal AK, Kondaiah P, Chakravarty AR. BODIPY-dipicolylamine complexes of platinum(II): X-ray structure, cellular imaging and organelle-specific near-IR light type-II PDT. Dalton Trans 2022; 51:3925-3936. [PMID: 35170587 DOI: 10.1039/d1dt03200c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Dipicolylamine (dpa) based platinum(II) complexes [Pt(L1-3)Cl]Cl (1-3), where L2 and L3 are green and red light BODIPY-tagged dpa ligands and L1 is a benzyl derivative of dpa, were synthesized and characterized and their in vitro cytotoxicity was studied. The perchlorate salt of complex 2 was structurally characterized. It showed a PtN3Cl core with a deformed square-planar geometry. At pH 7.2, complexes 2 and 3 showed strong absorption bands at 500 nm (ε ∼6.8 × 104 dm3 mol-1 cm-1) and 653 nm (ε ∼1.0 × 105 dm3 mol-1 cm-1) in a 1 : 1 (v/v) mixture of dimethyl sulfoxide and Dulbecco's phosphate-buffered saline (DMSO/DPBS), respectively. They displayed respective emission bands at 515 and 677 nm having fluorescence quantum yield values of 0.36 and 0.25. Complex 3 generated singlet oxygen, as evidenced from the 1,3-diphenylisobenzofuran titration experiments and mechanistic DNA photocleavage study. It showed high photocytotoxicity in red light (600-720 nm) with half-maximal inhibitory concentration (IC50) values of 1.73 and 2.67 μM in HeLa and A549 cells. The complexes showed significantly reduced chemo-PDT activity in a non-cancerous HPL1D cell line and in the dark. The 2',7'-dichlorofluorescein diacetate assay revealed reactive oxygen species-mediated type-II photodynamic therapy (PDT) activity. Cellular imaging of A549 cancer cells using complexes 2 and 3 revealed their preferential localization in mitochondria and endoplasmic reticulum. The annexin V-FITC/PI assay confirmed apoptotic cell damage. Cell cycle analysis indicated arrest in the G1 phase upon red light irradiation. Pt-DNA adduct formation was proposed from a DNA binding experiment with green light active complex 2 and 9-ethylguanine as a nucleobase from the mass spectral study.
Collapse
Affiliation(s)
- Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Srishti Gautam
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Apurba Kumar Pal
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| | - Paturu Kondaiah
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
25
|
The evolution of clinical guidelines for antimicrobial photodynamic therapy of skin. Photochem Photobiol Sci 2022; 21:385-395. [PMID: 35132604 PMCID: PMC8821777 DOI: 10.1007/s43630-021-00169-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/28/2021] [Indexed: 11/28/2022]
Abstract
Antimicrobial photodynamic therapy has become an important component in the treatment of human infection. This review considers historical guidelines, and the scientific literature to envisage what future clinical guidelines for treating skin infection might include. Antibiotic resistance, vertical and horizontal infection control strategies and a range of technologies effective in eradicating microbes without building up new resistance are described. The mechanism of action of these treatments and examples of their clinical use are also included. The research recommendations of NICE Guidelines on the dermatological manifestations of microbial infection were also reviewed to identify potential applications for PDT. The resistance of some microbes to antibiotics can be halted, or even reversed through the use of supplementary drugs, and so they are likely to persist as a treatment of infection. Conventional PDT will undoubtedly continue to be used for a range of skin conditions given existing healthcare infrastructure and a large evidence base. Daylight PDT may find broader antimicrobial applications than just Acne and Cutaneous Leishmaniasis, and Ambulatory PDT devices could become popular in regions where resources are limited or daylight exposure is not possible or inappropriate. Nanotheranostics were found to be highly relevant, and often include PDT, however, new treatments and novel applications and combinations of existing treatments will be subject to Clinical Trials.
Collapse
|
26
|
Laboe M, Lahiri J, Mohan T M N, Liang F, Levine BG, Beck WF, Dantus M. Linear and Nonlinear Optical Processes Controlling S 2 and S 1 Dual Fluorescence in Cyanine Dyes. J Phys Chem A 2021; 125:9770-9784. [PMID: 34747598 DOI: 10.1021/acs.jpca.1c05772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report on the changes in the dual fluorescence of two cyanine dyes IR144 and IR140 as a function of viscosity and probe their internal conversion dynamics from S2 to S1 via their dependence on a femtosecond laser pulse chirp. Steady-state and time-resolved measurements performed in methanol, ethanol, propanol, ethylene glycol, and glycerol solutions are presented. Quantum calculations reveal the presence of three excited states responsible for the experimental observations. Above the first excited state, we find an excited state, which we designate as S1', that relaxes to the S1 minimum, and we find that the S2 state has two stable configurations. Chirp-dependence measurements, aided by numerical simulations, reveal how internal conversion from S2 to S1 depends on solvent viscosity and pulse duration. By combining solvent viscosity, transform-limited pulses, and chirped pulses, we obtain an overall change in the S2/S1 population ratio of a factor of 86 and 55 for IR144 and IR140, respectively. The increase in the S2/S1 ratio is explained by a two-photon transition to a higher excited state. The ability to maximize the population of higher excited states by delaying or bypassing nonradiative relaxation may lead to the increased efficiency of photochemical processes.
Collapse
Affiliation(s)
- Maryann Laboe
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jurick Lahiri
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Nila Mohan T M
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Fangchun Liang
- Institute for Advanced Computational Science and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Benjamin G Levine
- Institute for Advanced Computational Science and Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Warren F Beck
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Marcos Dantus
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States.,Department of Physics and Astronomy, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
27
|
Paul S, Kundu P, Kondaiah P, Chakravarty AR. BODIPY-Ruthenium(II) Bis-Terpyridine Complexes for Cellular Imaging and Type-I/-II Photodynamic Therapy. Inorg Chem 2021; 60:16178-16193. [PMID: 34672556 DOI: 10.1021/acs.inorgchem.1c01850] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of multichromophoric ruthenium(II) complexes with the formulation [Ru(tpy-BODIPY)(tpy-R)]Cl2 (1-4), having a heteroleptic Ru(II)-bis-tpy (tpy = 4'-phenyl-2,2':6',2″-terpyridine) moiety covalently linked to a boron-dipyrromethene (BODIPY) pendant, have been prepared and characterized and their application as a phototherapeutic and photodetection agent in cancer therapy has been explored. Ligand L1 with a terpyridine-BODIPY moiety and complex 1 as its PF6 salt (1a) have been structurally characterized by a single-crystal X-ray diffraction study. Complex 1a has a distorted-octahedral RuN6 core with a Ru(II)-bis-terpyridine unit that is covalently linked to one photoactive BODIPY unit. The complexes exhibit strong absorbance near 502 nm (ε ≈ (3.7-7.8) × 104 M-1 cm-1) and high singlet oxygen sensitization ability, giving singlet oxygen quantum yield (ΦΔ) values ranging from 0.57 to 0.75 in DMSO. An emission-based study using complex 4 and Singlet Oxygen Sensor Green (SOSG) displays the formation of singlet oxygen inside the cells and also in the buffer medium upon light irradiation. DNA (pUC19) photocleavage experiments using ROS scavengers/stabilizers reveal photoinduced generation of singlet oxygen by a type-II process and of the superoxide anion radical by a type-I process. Complex 4 having a pendant biotin moiety as a cancer cell targeting group shows high photocytotoxicity with a remarkable phototherapeutic index (PI) value of >1400 in HeLa cancer cells with a low light dose activation (400-700 nm, 2.2 J cm-2). The complexes display reduced activity in noncancerous HPL1D cells. The emission property of the complexes is used for cellular imaging, thus making them suitable as next-generation theranostic PDT agents.
Collapse
|
28
|
Fudickar W, Roder P, Listek M, Hanack K, Linker T. Pyridinium Alkynylanthracenes as Sensitizers for Photodynamic Therapy. Photochem Photobiol 2021; 98:193-201. [PMID: 34719028 DOI: 10.1111/php.13554] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023]
Abstract
Photodynamic therapy (PDT) is a mild but effective method to treat certain types of cancer upon irradiation with visible light. Here, three isomeric methylpyridinium alkynylanthracenes 1o─p were evaluated as sensitizers for PDT. Upon irradiation with blue or green light, all three compounds show the ability to initiate strand breaks of plasmid DNA. The mayor species responsible for cleavage is singlet oxygen (1 O2 ) as confirmed by scavenging reagents. Only isomers 1m and 1p can be incorporated into HeLa cells, whereas isomer 1o cannot permeate through the membrane. While isomer 1m targets the cell nucleus, isomer 1p assembles in the cellular cytoplasm and impacts the cellular integrity. This is in accordance with a moderate toxicity of 1p in the dark, whereas 1m exhibits no dark toxicity. Both isomers are suitable as PDT reagents, with a CC50 of 3 μm and 75 nm, for 1p and 1m, respectively. Thus, derivative 1m, which can be easily synthesized, becomes an interesting candidate for cancer therapy.
Collapse
Affiliation(s)
- Werner Fudickar
- Department of Chemistry, University of Potsdam, Potsdam, Germany
| | - Phillip Roder
- Department of Chemistry, University of Potsdam, Potsdam, Germany
| | - Martin Listek
- Department of Biology, University of Potsdam, Potsdam, Germany
| | - Katja Hanack
- Department of Biology, University of Potsdam, Potsdam, Germany
| | - Torsten Linker
- Department of Chemistry, University of Potsdam, Potsdam, Germany
| |
Collapse
|
29
|
Recent Advances in Photodynamic Imaging and Therapy in Hepatobiliary Malignancies: Clinical and Experimental Aspects. Curr Oncol 2021; 28:4067-4079. [PMID: 34677263 PMCID: PMC8534451 DOI: 10.3390/curroncol28050345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/10/2023] Open
Abstract
The therapeutic and diagnostic modalities of light are well known, and derivative photodynamic reactions with photosensitizers (PSs), specific wavelengths of light exposure and the existence of tissue oxygen have been developed since the 20th century. Photodynamic therapy (PDT) is an effective local treatment for cancer-specific laser ablation in malignancies of some organs, including the bile duct. Although curability for extrahepatic cholangiocarcinoma is expected with surgery alone, patients with unresectable or remnant biliary cancer need other effective palliative therapies, including PDT. The effectiveness of PDT for cholangiocarcinoma has been reported experimentally or clinically, but it is not the standard option now due to problems with accompanied photosensitivity, limited access routes of irradiation, tumor hypoxia, etc. Novel derivative treatments such as photoimmunotherapy have not been applied in the field hepatobiliary system. Photodynamic diagnosis (PDD) has been more widely applied in the clinical diagnoses of liver malignancies or liver vascularization. At present, 5-aminolevulinic acid (ALA) and indocyanine green (ICG) dyes are mainly used as PSs in PDD, and ICG has been applied for detecting liver malignancies or vascularization. However, no ideal tools for combining both PDD and PDT for solid tumors, including hepatobiliary malignancies, have been clinically developed. To proceed with experimental and clinical trials, it is necessary to clarify the effective photosensitive drugs that are feasible for photochemical diagnosis and local treatment.
Collapse
|
30
|
Karpagam B, Sankarganesh M, Ravi L, Kesavan MP, Vinoth Kumar GG, Rajagopal G, Rajesh J. Synthesis, spectroscopic and crystal structure of nickel(II) complex of thiosemicarbazone basedSchiff base: Antimicrobial, anticancer and molecular docking studies. INORG CHEM COMMUN 2021. [DOI: 10.1016/j.inoche.2021.108850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
31
|
Das D, Noor A, Raza MK, Goswami TK. Co(II) complexes of curcumin and a ferrocene-based curcuminoid: a study on photo-induced antitumor activity. J Biol Inorg Chem 2021; 26:881-893. [PMID: 34550450 DOI: 10.1007/s00775-021-01899-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/29/2021] [Indexed: 02/08/2023]
Abstract
Co(II) complexes having a ferrocene-based curcuminoid (Fc-curH) ligand viz. [Co(L)2(Fc-cur)]ClO4 (1, 2), where L is phenanthroline base, namely, 1,10-phenanthroline (phen in 1) and dipyrido[3,2-a:2',3'-c]phenazine (dppz in 2) have been synthesized, characterized and evaluated as photochemotherapeutic agents in vitro. The corresponding Co(II) complexes of the naturally occurring polyphenol curcumin (curH), namely, [Co(L)2(cur)]ClO4 (3, 4), where L is phen (in 3) and dppz (in 4) were synthesized and their photo-induced anticancer activities compared with their ferrocene containing counterparts 1 and 2. The Co(II) acetylacetonato complex viz. [Co(phen)2(acac)]ClO4 (5) was structurally characterized through X-ray crystallography and used as control for cellular experiments. The Co(II) complexes having ferrocene-based curcuminoid are remarkably stable at physiological condition with higher lipophilicity compared to their curcumin analogues. The complexes display significant binding propensity to calf thymus (ct) DNA and human serum albumin (HSA). The complexes 1-4 display remarkable visible light induced cytotoxicity with the ferrocenyl analogues showing more phototoxic index (PI). The Co(II) curcumin complexes localize in the nucleus and mitochondria of A549 cells. The primary cell death mechanism is believed to be apoptotic in nature induced by light assisted generation of reactive oxygen species (ROS).Graphic abstract.
Collapse
Affiliation(s)
- Dhananjay Das
- Department of Chemistry, Gauhati University, Guwahati, 781014, Assam, India
| | - Aisha Noor
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, 110025, India
| | - Md Kausar Raza
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, 560012, India.
| | - Tridib K Goswami
- Department of Chemistry, Gauhati University, Guwahati, 781014, Assam, India.
| |
Collapse
|
32
|
Kim HJ, Montenegro D, Zhao J, Sparrow JR. Bisretinoids of the Retina: Photo-Oxidation, Iron-Catalyzed Oxidation, and Disease Consequences. Antioxidants (Basel) 2021; 10:antiox10091382. [PMID: 34573014 PMCID: PMC8467448 DOI: 10.3390/antiox10091382] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023] Open
Abstract
The retina and, in particular, retinal pigment epithelial cells are unusual for being encumbered by exposure to visible light, while being oxygen-rich, and also amassing photoreactive molecules. These fluorophores (bisretinoids) are generated as a byproduct of the activity of vitamin A aldehyde-the chromophore necessary for vision. Bisretinoids form in photoreceptor cells due to random reactions of two molecules of vitamin A aldehyde with phosphatidylethanolamine; bisretinoids are subsequently transferred to retinal pigment epithelial (RPE) cells, where they accumulate in the lysosomal compartment with age. Bisretinoids can generate reactive oxygen species by both energy and electron transfer, and they become photo-oxidized and photolyzed in the process. While these fluorescent molecules are accrued by RPE cells of all healthy eyes, they are also implicated in retinal disease.
Collapse
Affiliation(s)
- Hye Jin Kim
- Department of Ophthalmology, Columbia University Medical Center, 635 W., 165th Str., New York, NY 10032, USA; (H.J.K.); (D.M.); (J.Z.)
| | - Diego Montenegro
- Department of Ophthalmology, Columbia University Medical Center, 635 W., 165th Str., New York, NY 10032, USA; (H.J.K.); (D.M.); (J.Z.)
| | - Jin Zhao
- Department of Ophthalmology, Columbia University Medical Center, 635 W., 165th Str., New York, NY 10032, USA; (H.J.K.); (D.M.); (J.Z.)
| | - Janet R. Sparrow
- Department of Ophthalmology, Columbia University Medical Center, 635 W., 165th Str., New York, NY 10032, USA; (H.J.K.); (D.M.); (J.Z.)
- Department of Pathology and Cell Biology, Columbia University Medical Center, 635 W., 165th Str., New York, NY 10032, USA
- Correspondence: ; Tel.: +1-212-305-9944
| |
Collapse
|
33
|
Bilici K, Cetin S, Celikbas E, Yagci Acar H, Kolemen S. Recent Advances in Cyanine-Based Phototherapy Agents. Front Chem 2021; 9:707876. [PMID: 34249874 PMCID: PMC8263920 DOI: 10.3389/fchem.2021.707876] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 05/31/2021] [Indexed: 01/28/2023] Open
Abstract
Phototherapies, in the form of photodynamic therapy (PDT) and photothermal therapy (PTT), are very promising treatment modalities for cancer since they provide locality and turn-on mechanism for toxicity, both of which are critical in reducing off-site toxicity. Irradiation of photosensitive agents demonstrated successful therapeutic outcomes; however, each approach has its limitations and needs to be improved for clinical success. The combination of PTT and PDT may work in a synergistic way to overcome the limitations of each method and indeed improve the treatment efficacy. The development of single photosensitive agents capable of inducing both PDT and PTT is, therefore, extremely advantageous and highly desired. Cyanine dyes are shown to have such potential, hence have been very popular in the recent years. Luminescence of cyanine dyes renders them as phototheranostic molecules, reporting the localization of the photosensitive agent prior to irradiation to induce phototoxicity, hence allowing image-guided phototherapy. In this review, we mainly focus on the cyanine dye-based phototherapy of different cancer cells, concentrating on the advancements achieved in the last ten years.
Collapse
Affiliation(s)
- Kubra Bilici
- Department of Chemistry, Koc University, Istanbul, Turkey
| | - Sultan Cetin
- Department of Chemistry, Koc University, Istanbul, Turkey
| | - Eda Celikbas
- Department of Chemistry, Koc University, Istanbul, Turkey
| | - Havva Yagci Acar
- Department of Chemistry, Koc University, Istanbul, Turkey,Surface Science and Technology Center (KUYTAM), Koc University, Istanbul, Turkey,Graduate School of Materials Science and Engineering, Koc University, Istanbul, Turkey,*Correspondence: Havva Yagci Acar, ; Safacan Kolemen,
| | - Safacan Kolemen
- Department of Chemistry, Koc University, Istanbul, Turkey,Surface Science and Technology Center (KUYTAM), Koc University, Istanbul, Turkey,Boron and Advanced Materials Application and Research Center, Koc University, Istanbul, Turkey,TUPRAS Energy Center (KUTEM), Koc University, Istanbul, Turkey,*Correspondence: Havva Yagci Acar, ; Safacan Kolemen,
| |
Collapse
|
34
|
Lange N, Szlasa W, Saczko J, Chwiłkowska A. Potential of Cyanine Derived Dyes in Photodynamic Therapy. Pharmaceutics 2021; 13:818. [PMID: 34072719 PMCID: PMC8229084 DOI: 10.3390/pharmaceutics13060818] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022] Open
Abstract
Photodynamic therapy (PDT) is a method of cancer treatment that leads to the disintegration of cancer cells and has developed significantly in recent years. The clinically used photosensitizers are primarily porphyrin, which absorbs light in the red spectrum and their absorbance maxima are relatively short. This review presents group of compounds and their derivatives that are considered to be potential photosensitizers in PDT. Cyanine dyes are compounds that typically absorb light in the visible to near-infrared-I (NIR-I) spectrum range (750-900 nm). This meta-analysis comprises the current studies on cyanine dye derivatives, such as indocyanine green (so far used solely as a diagnostic agent), heptamethine and pentamethine dyes, squaraine dyes, merocyanines and phthalocyanines. The wide array of the cyanine derivatives arises from their structural modifications (e.g., halogenation, incorporation of metal atoms or organic structures, or synthesis of lactosomes, emulsions or conjugation). All the following modifications aim to increase solubility in aqueous media, enhance phototoxicity, and decrease photobleaching. In addition, the changes introduce new features like pH-sensitivity. The cyanine dyes involved in photodynamic reactions could be incorporated into sets of PDT agents.
Collapse
Affiliation(s)
- Natalia Lange
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (N.L.); (W.S.)
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (N.L.); (W.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| | - Agnieszka Chwiłkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland;
| |
Collapse
|
35
|
Short-term improvement of clinical parameters and microbial diversity in periodontitis patients following Indocyanine green-based antimicrobial photodynamic therapy: A randomized single-blind split-mouth cohort. Photodiagnosis Photodyn Ther 2021; 35:102349. [PMID: 34033939 DOI: 10.1016/j.pdpdt.2021.102349] [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: 03/17/2021] [Revised: 05/07/2021] [Accepted: 05/17/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Indocyanine green-mediated photodynamic therapy is effective against chronic periodontitis. Here, we evaluated the efficiency of indocyanine green-based adjunctive antimicrobial photodynamic therapy in non-surgical treatment of chronic periodontitis patients. METHODS Fifty-six periodontally involved teeth of 20 patients were treated with "scaling and root planing" (control group) or "scaling and root planing with indocyanine green-based (perio-green, 0.1 mg/ml) antimicrobial photodynamic therapy" (test group) using a split-mouth design. We performed clinical assessment of probing depth, gingival recession, clinical attachment loss, and other indices, while plaque samples were collected for microbiome analysis. RESULTS At baseline, periodontal depth and clinical attachment loss were significantly higher in the test group (p < 0.05), and at 1-month post-treatment, we observed a significant favorable reduction of both periodontal depth and clinical attachment loss in test and control sites, with lower means maintained at 3 months (p = 0.01 and p = 0.000, respectively). Additionally, analysis of variance showed significant improvements in periodontal depth and clinical attachment loss in the indocyanine green-antimicrobial photodynamic therapy group (p = 0.001), although not for clinical attachment loss in controls (p = 0.102). Moreover, a significant reduction was observed in test sites for bleeding on probing and residual pocket post-therapy (p = 0.04 and p = 0.0001 respectively). Furthermore, microbiome analysis identified Porphyromonons gingivalis, Treponema, and Tannerella in all samples with favorable changes in test sites (p = 0.07). CONCLUSION We observed a significant reduction in periodontal clinical parameters (periodontal depth and clinical attachment loss) in chronic periodontitis patients treated with antimicrobial photodynamic therapy as an adjunctive procedure to conventional scaling and root planing. This improvement was associated with periodontal pathogen reduction and increase in the healthy subgingival microbiome.
Collapse
|
36
|
Rodrigues CV, Johnson KR, Lombardi VC, Rodrigues MO, Sobrinho JA, de Bettencourt-Dias A. Photocytotoxicity of Thiophene- and Bithiophene-Dipicolinato Luminescent Lanthanide Complexes. J Med Chem 2021; 64:7724-7734. [PMID: 34018753 DOI: 10.1021/acs.jmedchem.0c01805] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
New thiophene-dipicolinato-based compounds, K2nTdpa (n = 1, 2), were isolated. Their anions are sensitizers of lanthanide ion (LnIII) luminescence and singlet oxygen generation (1O2). Emission in the visible and near-infrared regions was observed for the LnIII complexes with efficiencies (ϕLn) ϕEu = 33% and ϕYb = 0.31% for 1Tdpa2- and ϕYb = 0.07% for 2Tdpa2-. The latter does not sensitize EuIII emission. Fluorescence imaging of HeLa live cells incubated with K3[Eu(1Tdpa)3] indicates that the complex permeates the cell membrane and localizes in the mitochondria. All complexes generate 1O2 in solution with efficiencies (ϕO12) as high as 13 and 23% for the GdIII complexes of 1Tdpa2- and 2Tdpa2-, respectively. [Ln(nTdpa)3]3- (n = 1, 2) are phototoxic to HeLa cells when irradiated with UV light with IC50 values as low as 4.2 μM for [Gd(2Tdpa)3]3- and 91.8 μM for [Eu(1Tdpa)3]3-. Flow cytometric analyses indicate both apoptotic and necrotic cell death pathways.
Collapse
Affiliation(s)
- Carime V Rodrigues
- Department of Chemistry, University of Nevada, Reno, Reno, Nevada 89557, United States.,Laboratório de Inorgânica e Materiais, Instituto de Química, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Brasilia 70910-900 DF, Brazil
| | - Katherine R Johnson
- Department of Chemistry, University of Nevada, Reno, Reno, Nevada 89557, United States
| | - Vincent C Lombardi
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, Nevada 89557, United States
| | - Marcelo O Rodrigues
- Laboratório de Inorgânica e Materiais, Instituto de Química, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Brasilia 70910-900 DF, Brazil
| | - Josiane A Sobrinho
- Department of Chemistry, University of Nevada, Reno, Reno, Nevada 89557, United States
| | | |
Collapse
|
37
|
Kwon N, Kim H, Li X, Yoon J. Supramolecular agents for combination of photodynamic therapy and other treatments. Chem Sci 2021; 12:7248-7268. [PMID: 34163818 PMCID: PMC8171357 DOI: 10.1039/d1sc01125a] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/04/2021] [Indexed: 12/17/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising treatment for cancers such as superficial skin cancers, esophageal cancer, and cervical cancer. Unfortunately, PDT often does not have sufficient therapeutic benefits due to its intrinsic oxygen dependence and the limited permeability of irradiating light. Side effects from "always on" photosensitizers (PSs) can be problematic, and PDT cannot treat tumor metastases or recurrences. In recent years, supramolecular approaches using non-covalent interactions have attracted attention due to their potential in PS development. A supramolecular PS assembly could be built to maximize photodynamic effects and minimize side effects. A combination of two or more therapies can effectively address shortcomings while maximizing the benefits of each treatment regimen. Using the supramolecular assembly, it is possible to design a multifunctional supramolecular PS to exert synergistic effects by combining PDT with other treatment methods. This review provides a summary of important research progress on supramolecular systems that can be used to combine PDT with photothermal therapy, chemotherapy, and immunotherapy to compensate for the shortcomings of PDT, and it provides an overview of the prospects for future cancer treatment advances and clinical applications.
Collapse
Affiliation(s)
- Nahyun Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| | - Heejeong Kim
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| | - Xingshu Li
- College of Chemistry, State Key Laboratory of Photocatalysis on Energy and Environment, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Fuzhou University Fuzhou 350116 China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University Seoul 03760 Korea
| |
Collapse
|
38
|
Sahoo S, Raghavan A, Kumar A, Nandi D, Chakravarty AR. Biotin‐Appended Iron(III) Complexes of Curcumin for Targeted Photo‐Chemotherapy. Eur J Inorg Chem 2021. [DOI: 10.1002/ejic.202001174] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Somarupa Sahoo
- Department of Inorganic and Physical Chemistry Indian Institute of Science Bangalore 560012 Karnataka India
| | - Abinaya Raghavan
- Department of Biochemistry Indian Institute of Science Bangalore 560012 Karnataka India
| | - Arun Kumar
- Department of Inorganic and Physical Chemistry Indian Institute of Science Bangalore 560012 Karnataka India
| | - Dipankar Nandi
- Department of Biochemistry Indian Institute of Science Bangalore 560012 Karnataka India
| | - Akhil R. Chakravarty
- Department of Inorganic and Physical Chemistry Indian Institute of Science Bangalore 560012 Karnataka India
| |
Collapse
|
39
|
Babu B, Mack J, Nyokong T. An octabrominated Sn(iv) tetraisopropylporphyrin as a photosensitizer dye for singlet oxygen biomedical applications. Dalton Trans 2021; 49:9568-9573. [PMID: 32578634 DOI: 10.1039/d0dt01915a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Two novel Sn(iv) tetraisopropylphenylporphyrins have been synthesized to explore the effect of octabromination at the β-pyrrole positions on their photophysical properties and photodynamic activity. The lower energy Q band of an octabrominated complex lies at 675 nm well within the therapeutic window. The octabrominated dye has a relatively high singlet oxygen quantum yield of 0.78 in DMF and exhibits favorable photodynamic activity against MCF-7 cells with an IC50 value of 10.7 μM and a 5.74 log reduction value (5 μM) towards S. aureus under illumination at 660 nm for 60 min with a Thorlabs M660L3 LED (280 mW cm-2).
Collapse
Affiliation(s)
- Balaji Babu
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| | - John Mack
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| |
Collapse
|
40
|
Bhattacharyya A, Jameei A, Karande AA, Chakravarty AR. BODIPY-attached zinc(II) complexes of curcumin drug for visible light assisted photo-sensitization, cellular imaging and targeted PDT. Eur J Med Chem 2021; 220:113438. [PMID: 33915370 DOI: 10.1016/j.ejmech.2021.113438] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/28/2021] [Accepted: 04/02/2021] [Indexed: 12/13/2022]
Abstract
Boron-dipyrromethene (BODIPY) based photosensitizers as porphyrinoids and curcumin as natural product possess exciting photophysical features suitable for theranostic applications, namely, imaging and photodynamic therapy (PDT). Limited aqueous solubility and insufficient physiological stability, however, reduce their efficacy significantly. We have designed a novel strategy to deliver these two unusable cytotoxins simultaneously in cancer cells and herein, report the synthesis, characterization and imaging-assisted photocytotoxicity of three zinc(II) complexes containing N3-donor dipicolylamine (dpa) ligands (L1-3) and O,O-donor curcumin (Hcur) viz. [Zn(L1)(cur)]Cl (1), [Zn(L2)(cur)]Cl (2) and [Zn(L3)(cur)]Cl (3), where L2 and L3 have pendant fluorescent BODIPY and non-emissive di-iodo-BODIPY moieties. Metal chelation imparted remarkable biological stability (pH ∼7.4) to the respective ligands and induces significant aqueous solubility. These ternary complexes could act as replacements of the existing metalloporphyrin-based PDT photosensitizers as their visible-light photosensitizing ability is reinforced by the dual presence of blue light absorbing curcumin and green light harvesting BODIPY units. Complex 2 having emissive BODIPY unit L2 and curcumin, showed mitochondria selective localization in HeLa, MCF-7 cancer cells and complex 3, the di-iodinated analogue of complex 2, exhibited type-I/II PDT activity via inducing apoptosis through mitochondrial membrane disruption in cancer cells while being significantly nontoxic in dark and to the healthy cells.
Collapse
Affiliation(s)
- Arnab Bhattacharyya
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore, 560012, India
| | - Aida Jameei
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore, 560012, India
| | - Anjali A Karande
- Department of Biochemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore, 560012, India.
| | - Akhil R Chakravarty
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Sir C.V. Raman Avenue, Bangalore, 560012, India.
| |
Collapse
|
41
|
Sarkar T, Kumar A, Sahoo S, Hussain A. Mixed-Ligand Cobalt(III) Complexes of a Naturally Occurring Coumarin and Phenanthroline Bases as Mitochondria-Targeted Dual-Purpose Photochemotherapeutics. Inorg Chem 2021; 60:6649-6662. [PMID: 33855849 DOI: 10.1021/acs.inorgchem.1c00444] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The bioessential nature of cobalt and the rich photochemistry of its coordination complexes can be exploited to develop potential next-generation photochemotherapeutics. A series of six novel mixed-ligand cobalt(III) complexes of the formulation [Co(B)2(L)]ClO4 (1-6), where B is an N,N-donor phenanthroline base, namely, 1,10-phenanthroline (phen in 1 and 4), dipyrido[3,2-d:2',3'-f]quinoxaline (dpq in 2 and 5), and dipyrido[3,2-a:2',3'-c]phenazine (dppz in 3 and 6), and L is an O,O-donor dianionic ligand derived from catechol (1,2-dihydroxybenzene, cat2-, in 1-3) or esculetin (6,7-dihydoxycoumarin, esc2-, in 4-6), have been prepared and characterized, and their light-triggered cytotoxicity has been studied in cancer cells. The single-crystal X-ray diffraction structures of complexes 1 (as PF6- salt, 1a) and 2 show distorted octahedral geometries around the cobalt(III) center formed by the set of N4O2 donor atoms. The low-spin and 1:1 electrolytic complexes 1-6 display a d-d transition around 700 nm. Complexes 4-6 with a coordinated esc2- ligand additionally display a π → π* intraligand transition centered at 403 nm. Complexes 4-6 possessing a naturally occurring and photoactive esc2- ligand show high visible-light-triggered cytotoxicity against HeLa and MCF-7 cancer cells, yielding remarkably low micromolar IC50 values while being much less toxic under dark conditions. Control complexes 1-3 possessing the photoinactive cat2- ligand show significantly less cytotoxicity either in the presence of light or in the dark. The complex-induced cell death is apoptotic in nature caused by the formation of reactive oxygen species via a type 1 photoredox pathway. Fluorescence microscopy of HeLa cells treated with complex 6 reveals mitochondrial localization of the complex. A significant decrease in the dark toxicity of free esculetin and dppz base is observed upon coordination to cobalt(III). Complexes bind to calf-thymus DNA with significant affinity, but 6 binds with the greatest affinity. Complex 6 efficiently photocleaves supercoiled DNA to its nicked circular form when irradiated with visible light via a photoredox type 1 pathway involving hydroxyl radicals (HO•). Thus, complex 6 showing remarkable visible-light-triggered cytotoxicity but negligible toxicity in the dark is a good candidate for cancer photochemotherapy applications.
Collapse
Affiliation(s)
- Tukki Sarkar
- Department of Chemistry, Handique Girls' College, Guwahati 781001, Assam, India
| | - Arun Kumar
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Somarupa Sahoo
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Akhtar Hussain
- Department of Chemistry, Handique Girls' College, Guwahati 781001, Assam, India
| |
Collapse
|
42
|
Babu B, Mack J, Nyokong T. Photodynamic activity of Sn(IV) tetrathien-2-ylchlorin against MCF-7 breast cancer cells. Dalton Trans 2021; 50:2177-2182. [PMID: 33496304 DOI: 10.1039/d0dt03958f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new readily-synthesized Sn(iv) tetraarylchlorin with thien-2-yl substituents (SnC) has been prepared and fully characterized by various spectroscopic techniques and its photophysical and photochemical properties, such as the singlet oxygen quantum yield (ΦΔ), fluorescence quantum yield (ΦF), triplet lifetime (τT) and photostability, have been evaluated. SnC has an unusually high ΦΔ value of 0.89 in DMF. Studies on the photodynamic activity against MCF-7 breast cancer cells exhibited a very low IC50 value of 0.9 μM and high phototoxicity (dark versus light) indices of >27.8 after irradiation with a 660 nm Thorlabs LED (280 mW cm-2). The results demonstrate that Sn(iv) tetraarylchlorins of this type are suitable candidates for further in-depth PDT studies.
Collapse
Affiliation(s)
- Balaji Babu
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| | - John Mack
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| | - Tebello Nyokong
- Institute for Nanotechnology Innovation, Department of Chemistry, Rhodes University, Makhanda 6140, South Africa.
| |
Collapse
|
43
|
Li X, Zhao Y, Zhang T, Xing D. Mitochondria-Specific Agents for Photodynamic Cancer Therapy: A Key Determinant to Boost the Efficacy. Adv Healthc Mater 2021; 10:e2001240. [PMID: 33236531 DOI: 10.1002/adhm.202001240] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Mitochondria-targeted photodynamic therapy (Mt-PDT), which enables the photogenerated cytotoxic oxygen species with fatal oxidative damage to block mitochondrial functions, has been considered as a promising method to enhance the anticancer effectiveness. Aiming at the challenges of PDT, in the past few decades, numerous mitochondria-targeting molecular agents have been developed to boost the PDT efficacy via directly destroying the mitochondria or activating mitochondria-mediated cell death pathways. Herein, a review for recent advances of Mt-PDT is highlighted including: mitochondrial targeting design principles and strategies, therapeutic performance of mitochondria-targeted agents-mediated PDT as well as the agent-free Mt-PDT. In addition, it puts together the achievements of the combinatory mitochondria-anchoring PDT and other anticancer strategies, demonstrating the advantages provided by Mt-PDT. The existing challenges are discussed and future settlements for the development of mitochondria-specific agents are also forecasted.
Collapse
Affiliation(s)
- Xipeng Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Yu Zhao
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Tao Zhang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
- Guangdong Provincial Key Laboratory of Laser Life Science College of Biophotonics South China Normal University Guangzhou 510631 P. R. China
| |
Collapse
|
44
|
Hu T, Wang Z, Shen W, Liang R, Yan D, Wei M. Recent advances in innovative strategies for enhanced cancer photodynamic therapy. Theranostics 2021; 11:3278-3300. [PMID: 33537087 PMCID: PMC7847668 DOI: 10.7150/thno.54227] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/05/2020] [Indexed: 12/24/2022] Open
Abstract
Photodynamic therapy (PDT), a non-invasive therapeutic modality, has received increasing attention owing to its high selectivity and limited side effects. Although significant clinical research progress has been made in PDT, the breadth and depth of its clinical application have not been fully realized due to the limitations such as inadequate light penetration depth, non-targeting photosensitizers (PSs), and tumor hypoxia. Consequently, numerous investigations put their emphasis on innovative strategies to overcome the aforementioned limitations and enhance the therapeutic effect of PDT. Herein, up-to-date advances in these innovative methods for PDT are summarized by introducing the design of PS systems, their working mechanisms and application examples. In addition, current challenges of these innovative strategies for clinical application, and future perspectives on further improvement of PDT are also discussed.
Collapse
Affiliation(s)
- Tingting Hu
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Zhengdi Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Weicheng Shen
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Dan Yan
- Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Min Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
45
|
Bhattacharyya A, Jameei A, Saha R, Garai A, Karande AA, Chakravarty AR. BODIPY-linked cis-dichlorido zinc(ii) conjugates: the strategic design of organelle-specific next-generation theranostic photosensitizers. Dalton Trans 2021; 50:103-115. [DOI: 10.1039/d0dt03342a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cis-dichlorido Zn(ii)–BODIPY-based smart theranostic photosensitizers, as alternatives to Zn-porphyrins/phthalocyanines, show mitochondrion-targeted and imaging guided type-II photodynamic therapeutic activity.
Collapse
Affiliation(s)
- Arnab Bhattacharyya
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Aida Jameei
- Department of Biochemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Rupak Saha
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Aditya Garai
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Anjali A. Karande
- Department of Biochemistry
- Indian Institute of Science
- Bangalore 560012
- India
| | - Akhil R. Chakravarty
- Department of Inorganic and Physical Chemistry
- Indian Institute of Science
- Bangalore 560012
- India
| |
Collapse
|
46
|
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: 278] [Impact Index Per Article: 69.5] [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
|
47
|
Lee SY, Kim CY, Nam TG. Ruthenium Complexes as Anticancer Agents: A Brief History and Perspectives. Drug Des Devel Ther 2020; 14:5375-5392. [PMID: 33299303 PMCID: PMC7721113 DOI: 10.2147/dddt.s275007] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/03/2020] [Indexed: 12/28/2022] Open
Abstract
Platinum (Pt)-based anticancer drugs such as cisplatin have been used to treat various cancers. However, they have some limitations including poor selectivity and toxicity towards normal cells and increasing chemoresistance. Therefore, there is a need for novel metallo-anticancers, which has not been met for decades. Since the initial introduction of ruthenium (Ru) polypyridyl complex, a number of attempts at structural evolution have been conducted to improve efficacy. Among them, half-sandwich Ru-arene complexes have been the most prominent as an anticancer platform. Such complexes have clearly shown superior anticancer profiles such as increased selectivity toward cancer cells and ameliorating toxicity against normal cells compared to existing Pt-based anticancers. Currently, several Ru complexes are under human clinical trials. For improvement in selectivity and toxicity associated with chemotherapy, Ru complexes as photodynamic therapy (PDT), and photoactivated chemotherapy (PACT), which can selectively activate prodrug moieties in a specific region, have also been investigated. With all these studies on these interesting entities, new metallo-anticancer drugs to at least partially replace existing Pt-based anticancers are anticipated. This review covers a brief description of Ru-based anticancer complexes and perspectives.
Collapse
Affiliation(s)
- Sang Yeul Lee
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| | - Chul Young Kim
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| | - Tae-Gyu Nam
- Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Gyeonggi-do15588, Republic of Korea
| |
Collapse
|
48
|
Homsirikamol C, Suvanasuthi S, Viravaidya-Pasuwat K. Inclusion of IR-820 into Soybean-Phosphatides-Based Nanoparticles for Near-Infrared-Triggered Release and Endolysosomal Escape in HaCaT Keratinocytes at Insignificant Cytotoxic Level. Int J Nanomedicine 2020; 15:8717-8737. [PMID: 33192063 PMCID: PMC7654534 DOI: 10.2147/ijn.s267119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/22/2020] [Indexed: 01/15/2023] Open
Abstract
PURPOSE The degradation of drugs within endolysosomes has been widely addressed as a cause of poor bioavailability. One of the strategies to allow molecules to escape from a destructive fate is to introduce a photosensitizing moiety into a drug carrier enabling the permeabilization of endosomes and endolysosomes upon irradiation. This paper presents an alternative delivery nanosystem composed of cost-effective soybean phosphatides mixed with IR-820, a near-infrared (NIR) sensitizer, to load various active compounds and trigger an endolysosomal escape with a low cytotoxic effect. METHODS IR-820-incorporated phosphatides-based nanoparticles were formulated using a thin-film hydration method to encapsulate different molecular probes and a drug model. The nanoparticles were characterized in vitro using dynamic light scattering, transmission electron microscopy, as well as ultraviolet-visible and fluorescence spectroscopy techniques. The NIR-corresponding generation of the photochemical products, the content release, and the cytotoxicity toward the HaCaT keratinocyte cell line were evaluated. The cellular internalization and endolysosomal escape were monitored using a cytochemical marker and fluorescent probes with a colocalization analysis. RESULTS The IR-820-combined nanoparticles revealed the NIR-triggered changes in the singlet oxygen presence, nanoparticle architecture, and release rate without being cytotoxic. Additionally, the nanoplatform appeared to enhance cellular uptake of the macromolecules. The localization of the cytochemical marker and the colocalization analysis on the fluorescence signals of the encapsulated fluorophore and the lysosome-labeling reporter implied the transient endolysosomal escape of the cargo within the HaCaT cells after NIR irradiation. CONCLUSION The inclusion of IR-820 into a soybean-phosphatides base ingredient provides NIR responsiveness, particularly the endolysosomal escape of the payload, to the formulated nanoparticles, while preserving the beneficial properties as a drug carrier. This alternative delivery nanomedicine system has future potential to provide high bioavailability of cytosolic drugs utilizing time- and spatial-controllable NIR triggerability as well as the synergistic therapeutic effects with NIR-biomodulation.
Collapse
Affiliation(s)
- Chaiyarerk Homsirikamol
- Mammalian Cell Culture Laboratory, Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| | - Saroj Suvanasuthi
- Hair Diseases and Hair Transplantation Division, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Mammalian Cell Culture Laboratory, Biological Engineering Program, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
- Department of Chemical Engineering, Faculty of Engineering, King Mongkut’s University of Technology Thonburi, Bangkok, Thailand
| |
Collapse
|
49
|
Oliveira H, Correia P, Pereira AR, Araújo P, Mateus N, de Freitas V, Oliveira J, Fernandes I. Exploring the Applications of the Photoprotective Properties of Anthocyanins in Biological Systems. Int J Mol Sci 2020; 21:E7464. [PMID: 33050431 PMCID: PMC7589295 DOI: 10.3390/ijms21207464] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 12/13/2022] Open
Abstract
Due to their physical and chemical characteristics, anthocyanins are amongst the most versatile groups of natural compounds. Such unique signature makes these compounds a focus in several different areas of research. Anthocyanins have well been reported as bioactive compounds in a myriad of health disorders such as cardiovascular diseases, cancer, and obesity, among others, due to their anti-inflammatory, antioxidant, anti-diabetic, anti-bacterial, and anti-proliferative capacities. Such a vast number of action mechanisms may be also due to the number of structurally different anthocyanins plus their related derivatives. In this review, we highlight the recent advances on the potential use of anthocyanins in biological systems with particular focus on their photoprotective properties. Topics such as skin aging and eye degenerative diseases, highly influenced by light, and the action of anthocyanins against such damages will be discussed. Photodynamic Therapy and the potential role of anthocyanins as novel photosensitizers will be also a central theme of this review.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joana Oliveira
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal; (H.O.); (P.C.); (A.R.P.); (P.A.); (N.M.); (V.d.F.)
| | - Iva Fernandes
- LAQV, REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal; (H.O.); (P.C.); (A.R.P.); (P.A.); (N.M.); (V.d.F.)
| |
Collapse
|
50
|
Teng KX, Niu LY, Kang YF, Yang QZ. Rational design of a "dual lock-and-key" supramolecular photosensitizer based on aromatic nucleophilic substitution for specific and enhanced photodynamic therapy. Chem Sci 2020; 11:9703-9711. [PMID: 34094236 PMCID: PMC8162035 DOI: 10.1039/d0sc01122c] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Photosensitizing agents are essential for precise and efficient photodynamic therapy (PDT). However, most of the conventional photosensitizers still suffer from limitations such as aggregation-caused quenching (ACQ) in physiological environments and toxic side-effects on normal tissues during treatment, leading to reduced therapeutic efficacy. Thus, integrating excellent photophysical properties and accurate carcinoma selectivity in a photosensitizer system remains highly desired. Herein, a “dual lock-and-key” supramolecular photosensitizer BIBCl–PAE NPs for specific and enhanced cancer therapy is reported. BIBCl–PAE NPs are constructed by encapsulating a rationally designed glutathione (GSH)-activatable photosensitizer BIBCl in a pH-responsive diblock copolymer. In normal tissues, BIBCl is “locked” in the hydrophobic core of the polymeric micelles due to ACQ. Under the “dual key” activation of low pH and high levels of GSH in a tumor microenvironment, the disassembly of micelles facilitates the reaction of BIBCl with GSH to release water-soluble BIBSG with ideal biocompatibility, enabling the highly efficient PDT. Moreover, benefiting from the Förster resonance energy transfer effect of BIBSG, improved light harvesting ability and 1O2 production are achieved. In vitro and vivo experiments have demonstrated that BIBCl–PAE NPs are effective in targeting and inhibiting carcinoma. BIBCl–PAE NPs show superior anticancer efficiency relative to non-activatable controls. The “dual lock-and-key” supramolecular photosensitizers enable specific and enhanced photodynamic therapy (PDT).![]()
Collapse
Affiliation(s)
- Kun-Xu Teng
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University Beijing 100875 P. R. China
| | - Li-Ya Niu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University Beijing 100875 P. R. China
| | - Yan-Fei Kang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University Beijing 100875 P. R. China
| | - Qing-Zheng Yang
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University Beijing 100875 P. R. China
| |
Collapse
|