Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
J Am Chem Soc ; 144(23): 10647-10658, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35639988

ABSTRACT

Delivery of functional proteins into the intracellular space has been a challenging task that could lead to a myriad of therapeutic applications. We report herein a novel bioconjugation strategy for enzyme modification and selective delivery into cancer cells for lock-and-key-type activation of photosensitizers. Using a bifunctional linker containing a bis(bromomethyl)phenyl group and an o-phthalaldehyde moiety, it could induce cyclization of the peptide sequence Ac-NH-CRGDfC-CONH2 through site-specific dibenzylation with the two cysteine residues and further coupling with ß-galactosidase via the phthalaldehyde-amine capture reaction. This facile two-step one-pot procedure enabled the preparation of cyclic RGD-modified ß-galactosidase readily, which could be internalized selectively into αvß3 integrin-overexpressed cancer cells. Upon encountering an intrinsically quenched distyryl boron dipyrromethene-based photosensitizer conjugated with a galactose moiety through a self-immolative linker inside the cells, the extrinsic enzyme induced specific cleavage of the ß-galactosidic bond followed by self-immolation to release an activated derivative, thereby restoring the photodynamic activities and causing cell death effectively. The high specificity of this extrinsic enzyme-activated photosensitizing system was also demonstrated in vivo using nude mice bearing an αvß3 integrin-positive U87-MG tumor. The specific activation at the tumor site resulted in lighting up and complete eradication of the tumor upon laser irradiation, while by using the native ß-galactosidase, the effects were largely reduced. In contrast to the conventional activation using intrinsic enzymes, this extrinsic enzyme activatable approach can further minimize the nonspecific activation toward precisive photodynamic therapy.


Subject(s)
Photochemotherapy , Photosensitizing Agents , Animals , Cell Line, Tumor , Integrin beta3 , Mice , Mice, Nude , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , beta-Galactosidase
2.
Cancer Immunol Immunother ; 70(11): 3217-3233, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33821298

ABSTRACT

Photodynamic therapy (PDT) is a clinically approved treatment for various types of cancer. Besides killing the tumor cells directly, PDT has also been reported to trigger anti-tumor immunity. In our previous study, BAM-SiPc-based PDT was shown to induce immunogenic cell death on CT26 murine colon tumor cells in vitro. Using the BALB/c mouse animal model and a vascular-PDT (VPDT) approach, it could also eradicate tumor in ∼ 70% of tumor-bearing mice and elicit an anti-tumor immune response. In the present study, the serum obtained from the VPDT-cured mice was studied and found to possess various immunomodulatory properties. In in vitro studies, it stimulated cytokine secretions of IL-6 and C-X-C motif chemokine ligands 1-3 in CT26 cells through the NF-κB and MAPK pathways. The complement protein C5a boosted in the serum was shown to be involved in the process. The serum also induced calreticulin exposure on CT26 cells and activated dendritic cells. It contained CT26-targeting antibodies which, through the Fc region, induced macrophage engulfment of the tumor cells. In in vivo studies, inoculation of the serum-treated CT26 cells to mice demonstrated a retarded tumor growth with leukocytes, particularly T cells, attracted to the tumor site. In addition, the VPDT-cured mice showed different degrees of resistance against challenge of other types of murine tumor cells, for example, the breast tumor 4T1 and EMT6 cells.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Neoplasms, Experimental/blood , Neoplasms, Experimental/immunology , Photochemotherapy , Animals , Mice , Mice, Inbred BALB C
3.
Cancer Immunol Immunother ; 70(2): 485-495, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32839829

ABSTRACT

Photodynamic therapy (PDT) is an anti-tumor modality which employs three individually non-toxic substances, including photosensitizer, light and oxygen, to produce a toxic effect. Besides causing damage to blood vessels that supply oxygen and nutrients to the tumor and killing the tumor by a direct cytotoxic effect, PDT has also been known to trigger an anti-tumor immune response. For instance, our previous study showed that PDT with BAM-SiPc, a silicon(IV) phthalocyanine based-photosensitizer, can not only eradicate the mouse CT26 tumor cells in a Balb/c mouse model, but also protect the mice against further re-challenge of the tumor cells through an immunomodulatory mechanism. To understand more about the immune effect, the biochemical actions of BAM-SiPc-PDT on CT26 cells were studied in the in vitro system. It was confirmed that the PDT treatment could induce immunogenic necroptosis in the tumor cells. Upon treatment, different damage-associated molecular patterns were exposed onto the cell surface or released from the cells. Among them, calreticulin was found to translocate to the cell membrane through a pathway similar to that in chemotherapy. The activation of immune response was also demonstrated by an increase in the expression of different chemokines.


Subject(s)
Indoles/therapeutic use , Necroptosis/immunology , Organosilicon Compounds/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Animals , Cell Line, Tumor , Humans , Indoles/pharmacology , Isoindoles , Mice , Organosilicon Compounds/pharmacology , Photosensitizing Agents/pharmacology
4.
Mol Pharm ; 17(10): 3941-3951, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32931292

ABSTRACT

In cancer photodynamic therapy (PDT), a photosensitizer taken up by cancer cells can generate reactive oxygen species upon near-infrared light activation to induce cancer cell death. To increase PDT potency and decrease its adverse effect, one approach is to conjugate the photosensitizer with an antibody that specifically targets cancer cells. In the present study, IR700, a hydrophilic phthalocyanine photosensitizer, was conjugated to the humanized monoclonal antibody ARB102, which binds specifically cadherin-17 (CDH17 aka CA17), a cell surface marker highly expressed in gastrointestinal cancer to produce ARB102-IR700. Photoimmunotherapy (PIT) of gastrointestinal cancer cell lines was conducted by ARB102-IR700 treatment and near-infrared light irradiation. The results showed that ARB102-IR700 PIT could induce cell death in CDH17-positive cancer cells with high potency. In a co-culture model, CDH17-negative and CDH17-overexpressing SW480 cells were labeled with distinct fluorescent dyes and cultured together prior to PIT treatment. The results confirmed that ARB102-IR700 PIT could kill CDH17-positive cells specifically, while leaving the adjacent CDH17-negative cells unaffected. An in vivo efficacy study was conducted using a pancreatic adenocarcinoma AsPC-1 xenograft tumor model in nude mice. Fluorescence scanning indicated that ARB102-IR700 accumulated specifically in the tumor sites. To perform PIT, at 24 and 48 h postinjection, mice were irradiated with a 680 nm laser at the tumor site to activate the photosensitizer. It was shown that ARB102-IR700 PIT could inhibit tumor growth significantly. In summary, this study demonstrated that the novel ARB102-IR700 is a promising agent for PIT in gastrointestinal cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Cadherins/antagonists & inhibitors , Gastrointestinal Neoplasms/drug therapy , Photochemotherapy/methods , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cadherins/metabolism , Cell Culture Techniques , Cell Line, Tumor , Coculture Techniques , Female , Gastrointestinal Neoplasms/pathology , Humans , Infrared Rays , Injections, Intravenous , Mice , Tissue Distribution , Xenograft Model Antitumor Assays
5.
J Lipid Res ; 58(6): 1114-1131, 2017 06.
Article in English | MEDLINE | ID: mdl-28442498

ABSTRACT

PPARα has been known to play a pivotal role in orchestrating lipid, glucose, and amino acid metabolism via transcriptional regulation of its target gene expression during energy deprivation. Recent evidence has also suggested that PPARα is involved in bile acid metabolism, but how PPARα modulates the homeostasis of bile acids during fasting is still not clear. In a mechanistic study aiming to dissect the spectrum of PPARα target genes involved in metabolic response to fasting, we identified a novel mouse gene (herein named mL-STL for mouse liver-sulfotransferase-like) that shared extensive homology with the Sult2a subfamily of a superfamily of cytosolic sulfotransferases, implying its potential function in sulfonation. The mL-STL gene expressed predominantly in liver in fed state, but PPARα was required to sustain its expression during fasting, suggesting a critical role of PPARα in regulating the mL-STL-mediated sulfonation during fasting. Functional studies using recombinant His-tagged mL-STL protein revealed its narrow sulfonating activities toward 7α-hydroxyl primary bile acids, including cholic acid, chenodeoxycholic acid, and α-muricholic acid, and thus suggesting that mL-STL may be the major hepatic bile acid sulfonating enzyme in mice. Together, these studies identified a novel PPARα-dependent gene and uncovered a new role of PPARα as being an essential regulator in bile acid biotransformation via sulfonation during fasting.


Subject(s)
Bile Acids and Salts/metabolism , Cytosol/enzymology , PPAR alpha/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Amino Acid Sequence , Animals , Base Sequence , Biocatalysis , Biotransformation , Cloning, Molecular , DNA, Complementary/genetics , Down-Regulation , Fasting/metabolism , Liver/cytology , Male , Mice , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity , Sulfotransferases/chemistry
6.
Chemistry ; 23(65): 16505-16515, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28771861

ABSTRACT

Doxorubicin (Dox) was conjugated to a zinc(II) phthalocyanine (ZnPc) through an acid-cleavable hydrazone linker. This azido-containing conjugate was then anchored to the nanochannels of an alkyne-modified mesoporous silica nanoparticle (MSN) system via copper(I)-catalyzed azide-alkyne cycloaddition. An analogous nanosystem was also prepared by immobilization of a hydrazine-substituted ZnPc to the MSN followed by coupling with Dox. The release of Dox under acidic conditions was studied in phosphate-buffered saline. After internalization into human hepatocellular carcinoma HepG2 cells, these nanoparticles showed fluorescence not only for ZnPc, but also for Dox, suggesting that release of Dox was triggered by the acidic intracellular environment. The chemocytotoxic Dox together with singlet oxygen generated upon irradiation on the encapsulated ZnPc in these MSNs could kill the cells effectively. A synergistic cytotoxicity was suggested by a less-than-unity combination index. These nanoparticles function as both nanophotosensitizers for photodynamic therapy and as nanoplatforms for pH-controlled drug release.


Subject(s)
Doxorubicin/chemistry , Drug Carriers/chemistry , Indoles/chemistry , Nanoparticles/chemistry , Organometallic Compounds/chemistry , Silicon Dioxide/chemistry , Alkynes/chemistry , Cycloaddition Reaction , Doxorubicin/metabolism , Drug Liberation , Dynamic Light Scattering , Hep G2 Cells , Humans , Hydrogen-Ion Concentration , Isoindoles , Light , Microscopy, Confocal , Microscopy, Electron, Transmission , Photochemotherapy , Porosity , Singlet Oxygen/metabolism , Zinc Compounds
7.
Chem Commun (Camb) ; 59(23): 3471-3474, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36877479

ABSTRACT

A ß-galactosidase-responsive photosensitiser has been designed and synthesised. It contains a galactosyl substrate, a boron dipyrromethene-based photosensitising unit and a black hole quencher 2 connected via an AB2-type self-immolative linker. This novel photosensitiser can be selectively activated by the senescence-associated ß-galactosidase in senescent cells, leading to restoration in fluorescence emission and effective killing of the cells via photodynamic action.


Subject(s)
Galactosidases , Photosensitizing Agents , Photosensitizing Agents/pharmacology , beta-Galactosidase , Cell Line, Tumor , Cellular Senescence
8.
Apoptosis ; 17(4): 377-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22134530

ABSTRACT

Ribonucleases (RNases) are ubiquitously distributed nucleases that cleave RNA into smaller pieces. They are promising drugs for different cancers based on their concrete antitumor activities in vitro and in vivo. Here we report for the first time purification and characterization of a 14-kDa RNase, designated as RNase MC2, in the seeds of bitter gourd (Momordica charantia). RNase MC2 manifested potent RNA-cleavage activity toward baker's yeast tRNA, tumor cell rRNA, and an absolute specificity for uridine. RNase MC2 demonstrated both cytostatic and cytotoxic activities against MCF-7 breast cancer cells. Treatment of MCF-7 cells with RNase MC2 caused nuclear damage (karyorrhexis, chromatin condensation, and DNA fragmentation), ultimately resulting in early/late apoptosis. Further molecular studies unveiled that RNase MC2 induced differential activation of MAPKs (p38, JNK and ERK) and Akt. On the other hand, RNase MC2 exposure activated caspase-8, caspase-9, caspase-7, increased the production of Bak and cleaved PARP, which in turn contributed to the apoptotic response. In conclusion, RNase MC2 is a potential agent which can be exploited in the worldwide fight against breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/physiopathology , Caspases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Momordica charantia/enzymology , Ribonucleases/pharmacology , Amino Acid Sequence , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Caspase 7/genetics , Caspase 7/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Caspases/genetics , Cell Line, Tumor , Female , Humans , Mitogen-Activated Protein Kinases/genetics , Molecular Sequence Data , Momordica charantia/chemistry , Momordica charantia/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Ribonucleases/chemistry , Ribonucleases/genetics , Ribonucleases/isolation & purification , Seeds/chemistry , Seeds/enzymology , Sequence Alignment
9.
Chemistry ; 18(14): 4225-33, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22378352

ABSTRACT

A novel zinc(II) phthalocyanine conjugated with a short peptide with a nuclear localization sequence, Gly-Gly-Pro-Lys-Lys-Lys-Arg-Lys-Val, was synthesized by click chemistry and a standard Fmoc solid-phase peptide synthesis protocol. The conjugate was purified by HPLC and characterized with UV/Vis and high-resolution mass spectroscopic methods. Both this compound and its non-peptide-conjugated analogue are essentially non-aggregated in N,N-dimethylformamide and can generate singlet oxygen effectively with quantum yields (Φ(Δ)) of 0.84 and 0.81, respectively, relative to unsubstituted zinc(II) phthalocyanine (Φ(Δ) =0.56). Conjugation of the peptide sequence, however, can enhance the cellular uptake, efficiency in generating intracellular reactive oxygen species, and photocytotoxicity of the phthalocyanine-based photosensitizer against HT29 human colorectal carcinoma cells. The IC(50) value of the conjugate is as low as 0.21 µM. In addition, the conjugate shows an enhanced tumor-retention property in tumor-bearing nude mice. After 72 h post-injection, the dye concentration in the tumor was significantly higher than that in other organs. The results suggest that this phthalocyanine-peptide conjugate is a highly promising photosensitizer for photodynamic therapy.


Subject(s)
Formamides/chemistry , Indoles/chemistry , Indoles/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Peptides/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Click Chemistry , Dimethylformamide , Humans , Isoindoles , Mice , Photochemotherapy , Singlet Oxygen , Zinc Compounds
10.
BMC Cancer ; 12: 329, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22852552

ABSTRACT

BACKGROUND: Aldehyde dehydrogenases belong to a superfamily of detoxifying enzymes that protect cells from carcinogenic aldehydes. Of the superfamily, ALDH1A1 has gained most attention because current studies have shown that its expression is associated with human cancer stem cells. However, ALDH1A1 is only one of the 19 human ALDH subfamilies currently known. The purpose of the present study was to determine if the expression and activities of other major ALDH isozymes are associated with human ovarian cancer and ovarian cancer sphere cultures. METHODS: Immunohistochemistry was used to delineate ALDH isozyme localization in clinical ovarian tissues. Western Blot analyses were performed on lysates prepared from cancer cell lines and ovarian cancer spheres to confirm the immunohistochemistry findings. Quantitative reverse transcription-polymerase chain reactions were used to measure the mRNA expression levels. The Aldefluor® assay was used to measure ALDH activity in cancer cells from the four tumor subtypes. RESULTS: Immunohistochemical staining showed significant overexpression of ALDH1A3, ALDH3A2, and ALDH7A1 isozymes in ovarian tumors relative to normal ovarian tissues. The expression and activity of ALDH1A1 is tumor type-dependent, as seen from immunohistochemisty, Western blot analysis, and the Aldefluor® assay. The expression was elevated in the mucinous and endometrioid ovarian epithelial tumors than in serous and clear cell tumors. In some serous and most clear cell tumors, ALDH1A1 expression was found in the stromal fibroblasts. RNA expression of all studied ALDH isozymes also showed higher expression in endometrioid and mucinous tumors than in the serous and clear cell subtypes. The expression of ALDH enzymes showed tumor type-dependent induction in ovarian cancer cells growing as sphere suspensions in serum-free medium. CONCLUSIONS: The results of our study indicate that ALDH enzyme expression and activity may be associated with specific cell types in ovarian tumor tissues and vary according to cell states. Elucidating the function of the ALDH isozymes in lineage differentiation and pathogenesis may have significant implications for ovarian cancer pathophysiology.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Isoenzymes/metabolism , Ovarian Neoplasms/enzymology , Clinical Enzyme Tests/methods , Female , Humans , Immunohistochemistry , Ovarian Neoplasms/pathology , Spheroids, Cellular/enzymology , Tumor Cells, Cultured
11.
Phytother Res ; 26(5): 734-42, 2012 May.
Article in English | MEDLINE | ID: mdl-22072524

ABSTRACT

Breast cancer is conventionally treated by surgery and radiotherapy, with adjuvant chemotherapy and hormonotherapy as supplementary treatments. However, such treatments are associated with adverse side effects and drug resistance. In this study, Pheophorbide a (Pa), a photosensitizer isolated from Scutelleria barbata, was analysed for its antiproliferative effect on human breast tumour cells. The IC (inhibitory concentration)(50) of the combined treatment of Pa and photodynamic therapy (Pa-PDT) on human breast tumour MCF-7 cells was 0.5 µm. Mechanistic studies in MCF-7 cells demonstrated that Pa was localized in the mitochondria, and reactive oxygen species were found to be released after Pa-PDT. Apoptosis was the major mechanism responsible for the tumour cell death, and mitochondrial membrane depolarization and cytochrome c release highlighted the role of mitochondria in the apoptotic mechanism. Up-regulation of tumour suppressor protein p53, cleavage of caspase-9 and poly (ADP-ribose) polymerase suggested that the caspase-dependent pathway was induced, while the release of apoptosis-inducing factors demonstrated that the apoptosis was also mediated by the caspase-independent mechanism. In vivo study using the mouse xenograft model showed a significant inhibition of MCF-7 tumour growth by Pa-PDT. Together, the results of this study provide a basis for understanding and developing Pa-PDT as a cure for breast cancer.


Subject(s)
Apoptosis/drug effects , Chlorophyll/analogs & derivatives , Photochemotherapy , Photosensitizing Agents/pharmacology , Plant Extracts/pharmacology , Scutellaria/chemistry , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 9/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorophyll/pharmacology , Chlorophyll/therapeutic use , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Photosensitizing Agents/therapeutic use , Plant Extracts/therapeutic use , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
12.
ACS Appl Mater Interfaces ; 14(13): 14903-14915, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-35333503

ABSTRACT

The delivery and activation of photosensitizers in a specific manner is crucial in photodynamic therapy. For an antitumoral application, it can confine the photodynamic action on the cancer cells, thereby enhancing the treatment efficacy and reducing the side effects. We report herein a novel supramolecular photosensitizing nanosystem that can be specifically activated in cancer cells and tumors that overexpress epidermal growth factor receptor (EGFR). It involves the self-assembly of the amphiphilic host-guest complex of a ß-cyclodextrin-conjugated phthalocyanine-based photosensitizer (Pc-CD) and a ferrocene-substituted poly(ethylene glycol) (Mn = 2000) (Fc-PEG) in aqueous media. The resulting nanosystem Pc-CD@Fc-PEG with a hydrodynamic diameter of 124-147 nm could not emit fluorescence and generate reactive oxygen species due to the self-quenching effect and the ferrocene-based quencher. Upon interactions with molecules of adamantane substituted with an EGFR-targeting peptide (Ad-QRH*) in water and in EGFR-positive HT29 and A431 cells, the ferrocene guest species were displaced, resulting in disassembly of the nanoparticles and restoration of these photoactivities. The half-maximal inhibitory concentration values were down to 1.24 µM (for HT29 cells). The nanosystem Pc-CD@Fc-PEG could also be activated in an Ad-QRH*-treated HT29 tumor in nude mice, leading to increased intratumoral fluorescence intensity and effective eradication of the tumor upon laser irradiation. The results showed that this two-step supramolecular approach can actualize site-specific photosensitization and minimize nonspecific phototoxicity in a general photodynamic treatment.


Subject(s)
Cyclodextrins , Nanoparticles , Photochemotherapy , Animals , Cell Line, Tumor , Mice , Mice, Nude , Nanoparticles/chemistry , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
13.
Chemistry ; 17(27): 7569-77, 2011 Jun 27.
Article in English | MEDLINE | ID: mdl-21598326

ABSTRACT

A series of silicon(IV) phthalocyanines linked to two permethylated ß-cyclodextrin moieties through different spacers at the axial positions have been synthesized and characterized. The effects of these spacers on the photophysical properties and in vitro photodynamic activities have also been examined. Having two bulky hydrophilic substituents, all of these compounds are soluble and essentially nonaggregated in DMF and even in aqueous media. The fluorescence and singlet oxygen quantum yields in DMF are lower for the analogue with the shortest separation between the amino group in the spacer and the phthalocyanine ring. The fluorescence quantum yield of this compound increases in water probably due to protonation of the amino group, which inhibits the reductive quenching process. This series of compounds also exhibit photocytotoxicity toward HT29 human colon adenocarcinoma and HepG2 human hepatocarcinoma cells with IC(50) values in the range of 0.04-1.32 µM. The analogue with an α,ω-aminohydroxypentyl linker shows the highest potency, which can be ascribed to its high cellular uptake and high efficiency in generating intracellular reactive oxygen species. This compound also shows preferential localization in the lysosome, induces cell death mainly through apoptosis, and inhibits the growth of tumor in vivo. The results suggest that it is a promising photosensitizer for photodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/chemistry , Organosilicon Compounds/chemistry , Photosensitizing Agents/pharmacology , beta-Cyclodextrins/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Quantum Theory , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured
14.
Org Biomol Chem ; 9(20): 7028-32, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21858322

ABSTRACT

Treatment of 4,5-bis[4-(methoxycarbonyl)phenoxy]phthalonitrile and 4,5-bis[3,5-bis(methoxycarbonyl)phenoxy]phthalonitrile with an excess of 1,3-diiminoisoindoline in the presence of Zn(OAc)(2)·2H(2)O and 1,8-diazabicyclo[5.4.0]undec-7-ene in triethylene glycol monomethyl ether or polyethylene glycol monomethyl ether (with an average molecular weight of 550) led to "3 + 1" mixed cyclisation and transesterification in one pot, affording the corresponding di-ß-substituted zinc(II) phthalocyanines in 7-23% yield. As shown by absorption spectroscopy, these compounds were essentially non-aggregated in N,N-dimethylformamide and could generate singlet oxygen effectively. The singlet oxygen quantum yields (Φ(Δ) = 0.53-0.57) were comparable with that of the unsubstituted zinc(II) phthalocyanine (Φ(Δ) = 0.56). These compounds in Cremophor EL emulsions also exhibited photocytotoxicity against HT29 human colorectal adenocarcinoma and HepG2 human hepatocarcinoma cells with IC(50) values in the range of 0.25-3.72 µM. The analogue with four triethylene glycol chains was the most potent photosensitiser and localised preferentially in the mitochondria of HT29 cells. The bis(polyethylene glycol)-counterpart could form surfactant-free nanoparticles both in water and in the culture medium. The hydrodynamic radii, as determined by dynamic laser light scattering, ranged from 6.3 to 79.8 nm depending on the preparation methods and conditions. The photocytotoxicity of these nanoparticles (IC(50) = 0.43-0.56 µM) was comparable with that of the Cremophor EL-formulated system (IC(50) = 0.34 µM).


Subject(s)
Indoles/chemical synthesis , Organometallic Compounds/chemical synthesis , Zinc/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Esterification , Humans , Indoles/pharmacology , Isoindoles , Molecular Structure , Nanoparticles/chemistry , Organometallic Compounds/pharmacology , Structure-Activity Relationship
15.
J Med Chem ; 64(20): 15461-15476, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34662121

ABSTRACT

Targeted delivery of photosensitizers using hydrophilic and tumor-directing carriers and site-specific activation of their photocytotoxicity are two common strategies to enhance the specificity of anticancer photodynamic therapy. We report herein a novel supramolecular bio-orthogonal approach to integrate these two functions. A ß-cyclodextrin-substituted aza-boron-dipyrromethene-based photosensitizer was first complexed with a ferrocene-substituted black-hole quencher to inhibit its photosensitizing ability. Upon encountering the adamantane moieties that had been delivered to target cancer cells through specific binding of the conjugated peptide to the overexpressed epidermal growth factor receptor, the ferrocene-based guest species were displaced due to the stronger binding interactions between ß-cyclodextrin and adamantane, thereby restoring the photodynamic activity of the photosensitizer. Hence, this two-step process enabled targeted delivery and site-specific activation of the photosensitizer, as demonstrated through a series of experiments in aqueous media, in a range of cancer cell lines and in tumor-bearing nude mice.


Subject(s)
Antineoplastic Agents/pharmacology , Aza Compounds/pharmacology , Boron Compounds/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , beta-Cyclodextrins/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aza Compounds/chemistry , Boron Compounds/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Hydrophobic and Hydrophilic Interactions , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Structure-Activity Relationship , beta-Cyclodextrins/chemistry
16.
J Med Chem ; 64(4): 2064-2076, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33577327

ABSTRACT

A facile procedure for in situ peptide cyclization and phthalocyanine conjugation was developed by utilizing a bifunctional linker incorporated with a bis(bromomethyl)benzene unit and a cyclopentadiene moiety. These functional groups facilitated the nucleophilic substitution with the two cysteine residues of the linear peptides followed by the Diels-Alder reaction with the maleimide moiety attached to a zinc(II) phthalocyanine. With this approach, three cyclic peptide-phthalocyanine conjugates were prepared in 20-26% isolated yield via a one-pot procedure. One of the conjugates containing a cyclic form of the epidermal growth factor receptor (EGFR)-binding peptide sequence CMYIEALDKYAC displayed superior features as an advanced photosensitizer. It showed preferential uptake by two EGFR-positive cancer cell lines (HT29 and HCT116) compared with two EGFR-negative counterparts (HeLa and HEK293), resulting in significantly higher photocytotoxicity. Intravenous administration of this conjugate into HT29 tumor-bearing nude mice resulted in selective localization in tumor and effective inhibition of tumor growth upon photodynamic treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , ErbB Receptors/metabolism , Indoles/therapeutic use , Peptides, Cyclic/therapeutic use , Photosensitizing Agents/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , Female , HEK293 Cells , Humans , Indoles/chemical synthesis , Indoles/metabolism , Indoles/radiation effects , Light , Mice, Inbred BALB C , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Peptides, Cyclic/radiation effects , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/metabolism , Photosensitizing Agents/radiation effects , Precision Medicine
17.
J Med Chem ; 64(23): 17455-17467, 2021 12 09.
Article in English | MEDLINE | ID: mdl-34846143

ABSTRACT

Two dual stimuli-activated photosensitizers were developed, in which two or three glutathione (GSH)-responsive 2,4-dinitrobenzenesulfonate (DNBS)-substituted zinc(II) phthalocyanine units were connected via one or two cathepsin B-cleavable Gly-Phe-Leu-Gly peptide linker(s). These dimeric and trimeric phthalocyanines were fully quenched in the native form due to the photoinduced electron transfer to the DNBS substituents and the self-quenching of the phthalocyanine units. In the presence of GSH and cathepsin B, or upon internalization into A549 and HepG2 cancer cells, these probes were activated through the release of free phthalocyanine units. The intracellular fluorescence intensity was increased upon post-incubation with GSH ester or reduced upon pre-treatment with a cathepsin B inhibitor. Upon light irradiation, these photosensitizers became highly cytotoxic with IC50 values of 0.21-0.39 µM. The photocytotoxicity was also dependent on the intracellular GSH and cathepsin B levels. The results showed that these conjugates could serve as smart photosensitizers for targeted photodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biopolymers/metabolism , Cathepsin B/metabolism , Glutathione/metabolism , Isoindoles/metabolism , Photochemotherapy , Photosensitizing Agents/pharmacology , Cell Line, Tumor , Fluorescence , Humans
18.
Biomater Sci ; 10(1): 189-201, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34817474

ABSTRACT

A series of glutathione (GSH)-responsive polydopamine (PDA) nanoparticles (NPs) were prepared using a disulfide-linked dopamine dimer as starting material, of which the size could be tuned systematically by adjusting the amount of ammonia solution used. Molecules of a phthalocyanine (Pc)-based photosensitiser and an epidermal growth factor receptor (EGFR)-targeting peptide were then sequentially immobilised on the surface of the NPs through coupling with the surface functionalities of PDA. The immobilised Pc molecules in the resulting nanosystem were photodynamically inactive due to the strong self-quenching effect and the quenching by the PDA core. Upon exposure to GSH in phosphate-buffered saline or EGFR-positive cancer cells, namely A549 and A431 cells, the NPs were disassembled through cleavage of the disulfide linkages to release the Pc molecules, thereby restoring their fluorescence emission and singlet oxygen generation. The NPs with the smallest size (ca. 200 nm in diameter) exhibited the highest cellular uptake and high photocytotoxicity with IC50 values as low as 0.05 µM based on Pc. These NPs could also accumulate and be activated in the tumour of A431 tumour-bearing nude mice, lighting up the tumour with fluorescence over a period of 72 h and completely eradicating the tumour through laser irradiation for 10 min (675 nm, 20 J cm-2). The results suggest that these biodegradable and versatile PDA-based NPs can serve as a promising nanoplatform for fabrication of advanced photosensitisers for targeted photodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles , Photochemotherapy , Photosensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Glutathione , Indoles , Mice , Mice, Nude , Polymers
19.
Biomater Sci ; 9(23): 7832-7837, 2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34726672

ABSTRACT

We report herein a one-pot approach to cyclise a tumour-targeting peptide and conjugate it on the surface of red blood cells loaded with a boron dipyrromethene-based photosensitiser using a bifunctional linker consisting of a bis(bromomethyl)phenyl unit and an ortho-phthalaldehyde unit. This cell-based photosensitiser with surface modification with cyclic RGD peptide moieties can selectively bind against the αvß3 integrin-overexpressed cancer cells, leading to enhanced photocytotoxicity. The results demonstrate that this facile strategy is effective for live-cell surface modification for a wide range of applications.


Subject(s)
Neoplasms , Photochemotherapy , Cell Line, Tumor , Erythrocytes , Humans , Neoplasms/drug therapy , Peptides , Photosensitizing Agents/therapeutic use
20.
J Cell Biochem ; 109(1): 74-81, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19885858

ABSTRACT

Antiquitin is an aldehyde dehydrogenase involved in the catabolism of lysine. Mutations of antiquitin have been linked with the disease pyridoxine-dependent seizures. While it is well established that lysine metabolism takes place in the mitochondrial matrix, evidence for the mitochondrial localization of antiquitin has been lacking. In the present study, the subcellular localization of antiquitin was investigated using human embryonic kidney HEK293 cells. Three different approaches were used. First, confocal microscopic analysis was carried out on cells transiently transfected with fusion constructs containing enhanced green fluorescent protein with different lengths of antiquitin based on the different potential start codons of translation. Second, immunofluorescence staining was used to detect the localization of antiquitin directly in the cells. Third, subcellular fractionation was carried out and the individual fraction was analyzed for the presence of antiquitin by Western blot and flow cytometric analyses. All the results showed that antiquitin was present not only in the cytosol but also in the mitochondria.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Mitochondria/enzymology , Blotting, Western , Cell Line , Cell Separation , Cytosol/enzymology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL