Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 13 de 13
1.
Photochem Photobiol ; 96(3): 570-580, 2020 05.
Article En | MEDLINE | ID: mdl-32104926

Active targeting strategies are currently being extensively investigated in order to enhance the selectivity of photodynamic therapy. The aim of the present research was to evaluate whether the external decoration of nanopolymeric carriers with targeting peptides could add more value to a photosensitizer formulation and increase antitumor therapeutic efficacy and selectivity. To this end, we assessed PLGA-PLA-PEG nanoparticles (NPs) covalently attached to a hydrophilic photosensitizer 5-[4-azidophenyl]-10,15,20-tri-(N-methyl-4-pyridinium)porphyrinato zinc (II) trichloride (ZnTriMPyP) and also to c(RGDfK) peptides, in order to target αv ß3 integrin-expressing cells. In vitro phototoxicity investigations showed that the ZnTriMPyP-PLGA-PLA-PEG-c(RGDfK) nanosystem is effective at submicromolar concentrations, is devoid of dark toxicity, successfully targets αv ß3 integrin-expressing cells and is 10-fold more potent than related nanosystems where the PS is occluded instead of covalently bound.


Drug Carriers , Nanoparticles , Neoplasms/drug therapy , Oligopeptides/chemistry , Photochemotherapy , Photosensitizing Agents/pharmacology , Polymers/chemistry , Cell Line, Tumor , Humans , Integrins/drug effects , Kinetics , Photosensitizing Agents/therapeutic use , Singlet Oxygen/chemistry , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
2.
Bioconjug Chem ; 29(11): 3516-3521, 2018 11 21.
Article En | MEDLINE | ID: mdl-30352511

Antibody-drug conjugates (ADC) are spearheading vectorized chemotherapy against cancer, with 4 FDA-approved ADCs and 79 in clinical trials. However, most ADCs are produced using a stochastic bioconjugation method, target hematological cancers, and are derived from a full immunoglobulin-G (IgG). These factors limit their efficacy, especially against solid tumors which remain difficult to treat. Here we report the site-specific conjugation of a single auristatin derivative onto an engineered anti-HER2 single chain fragment variable (scFv) of the trastuzumab antibody, generating new scFv-drug conjugates (SDC). Two cysteines were judiciously incorporated at the beginning of the scFv hexahistidine tag, in order to allow controlled bioconjugation of a heterobifunctional linker including a second generation maleimide (SGM), either cleavable (for monomethyl auristatin E) or noncleavable (for monomethyl auristatin F). Our data indicated that both SDCs conserved their affinity to HER2 in comparison to the native scFv, and were efficiently able to kill in vitro HER2-positive SK-BR-3 cells at subnanomolar concentrations (EC50 of 0.68 nM and 0.32 nM). No effect was observed on HER2-negative MCF-7 cells. Ours results showed efficient targeting of site-specific SDCs against HER2-positive breast cancer cells. This work represents a first important step in the design of more effective small conjugates, paving the way for future in vivo translation to evaluate their full potential.


Aminobenzoates/chemistry , Breast Neoplasms/drug therapy , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Maleimides/chemistry , Oligopeptides/chemistry , Receptor, ErbB-2/drug effects , Single-Chain Antibodies/chemistry , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Immunoconjugates/therapeutic use , Immunologic Factors/therapeutic use , Protein Engineering , Trastuzumab/chemistry , Trastuzumab/immunology
3.
Org Biomol Chem ; 16(11): 1882-1889, 2018 03 14.
Article En | MEDLINE | ID: mdl-29473076

Herein we describe the synthesis and evaluation of four novel HER2-targeting, cathepsin B-sensitive antibody-drug conjugates bearing a monomethylauristatin E (MMAE) cytotoxic payload, constructed via the conjugation of cleavable linkers to trastuzumab using a site-specific bioconjugation methodology. These linkers vary by both cleavable trigger motif and hydrophilicity, containing one of two cathepsin B sensitive dipeptides (Val-Cit and Val-Ala), and engendered with either hydrophilic or hydrophobic character via application of a PEG12 spacer. Through evaluation of physical properties, in vitro cytotoxicity, and receptor affinity of the resulting antibody-drug conjugates (ADCs), we have demonstrated that while both dipeptide triggers are effective, the increased hydrophobicity of the Val-Ala pair limits its utility within this type of linker. In addition, while PEGylation augments linker hydrophilicity, this change does not translate to more favourable ADC hydrophilicity or potency. While all described structures demonstrated excellent and similar in vitro cytotoxicity, the ADC with the ValCitPABMMAE linker shows the most promising combination of in vitro potency, structural homogeneity, and hydrophilicity, warranting further evaluation into its therapeutic potential.


Antineoplastic Agents, Immunological/chemistry , Cathepsin B/metabolism , Immunoconjugates/chemistry , Oligopeptides/chemistry , Receptor, ErbB-2/metabolism , Trastuzumab/chemistry , Antineoplastic Agents, Immunological/chemical synthesis , Antineoplastic Agents, Immunological/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Chemistry Techniques, Synthetic/methods , Drug Delivery Systems , Humans , Hydrophobic and Hydrophilic Interactions , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Trastuzumab/pharmacology
4.
Bioconjug Chem ; 29(1): 176-181, 2018 01 17.
Article En | MEDLINE | ID: mdl-29216717

Exploitation of photosensitizers as payloads for antibody-based anticancer therapeutics offers a novel alternative to the small pool of commonly utilized cytotoxins. However, existing bioconjugation methodologies are incompatible with the requirement of increased antibody loading without compromising antibody function, stability, or homogeneity. Herein, we describe the first application of dendritic multiplier groups to allow the loading of more than 4 porphyrins to a full IgG antibody in a site-specific and highly homogeneous manner. Photophysical evaluation of UV-visible absorbance and singlet oxygen quantum yields highlighted porphyrin-dendron 14 as the best candidate for bioconjugation; with subsequent bioconjugation producing a HER2-targeted therapeutic with average loading ratios of 15.4:1. In vitro evaluation of conjugate 18 demonstrated a nanomolar photocytotoxic effect in a target cell line, which overexpresses HER2, with no observed photocytotoxicity at the same concentration in a control cell line which expresses native HER2 levels, or in the absence of irradiation with visible light.


Dendrimers/chemistry , Immunoconjugates/chemistry , Immunoglobulin G/chemistry , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Trastuzumab/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dendrimers/pharmacology , Drug Delivery Systems , Humans , Immunoconjugates/pharmacology , Immunoglobulin G/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Receptor, ErbB-2/analysis , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology
5.
Eur J Med Chem ; 142: 393-415, 2017 Dec 15.
Article En | MEDLINE | ID: mdl-28911823

The design of innovative anticancer chemotherapies with superior antitumor efficacy and reduced toxicity continues to be a challenging endeavor. Recently, the success of Adcetris® and Kadcyla® made antibody-drug conjugates (ADCs) serious contenders to reach the envied status of Paul Ehrlich's "magic bullet". However, ADCs classically target overexpressed and internalizing antigens at the surface of cancer cells, and in solid tumors are associated with poor tumor penetration, insufficient targeting in heterogeneous tumors, and appearance of several resistance mechanisms. In this context, alternative non-internalizing ADCs and prodrugs have been developed to circumvent these limitations, in which the drug can be selectively released by an extracellular stimulus in the tumor microenvironment. Each strategy and method of activation will be discussed as potential alternatives to internalizing ADCs for cancer therapy.


Antineoplastic Agents, Immunological/therapeutic use , Immunoconjugates/therapeutic use , Neoplasms/drug therapy , Prodrugs/therapeutic use , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , Drug Delivery Systems , Drug Discovery , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/pharmacokinetics , Immunoconjugates/pharmacology , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Prodrugs/pharmacology
6.
Photochem Photobiol Sci ; 16(8): 1260-1267, 2017 Aug 09.
Article En | MEDLINE | ID: mdl-28636039

Fluorescence is an essential imaging modality for labelling and visualising cells and sub-cellular structures. Multicolour labelling is especially challenging due to differences in physicochemical and photophysical behaviour of structurally unrelated fluorophores in the heterogeneous environments found in sub-cellular compartments. Herein, we report the conjugation of three azide-bearing BODIPYs with similar core structures but widely different emission wavelengths (green, red and NIR) to tyrosine residues of a model globular protein (BSA) via a common linking methodology. The resulting BODIPY-BSA conjugates have demonstrated multi-wavelength fluorescence emission for biological applications. Fluorescence imaging was performed in HeLa cells through live cell confocal microscopy imaging, with good intracellular location visualisation observed.


Boron Compounds/chemistry , Serum Albumin, Bovine/chemistry , Tyrosine/chemistry , Amino Acid Sequence , Animals , Boron Compounds/metabolism , Cattle , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Infrared Rays , Microscopy, Confocal , Serum Albumin, Bovine/metabolism , Spectrophotometry , Tyrosine/metabolism
7.
J Photochem Photobiol B ; 163: 374-84, 2016 Oct.
Article En | MEDLINE | ID: mdl-27619739

Duramycin, through binding with phosphatidylethanolamine (PE), has been shown to be a selective molecular probe for the targeting and imaging of cancer cells. Photodynamic therapy aims to bring about specific cytotoxic damage to tumours through delivery of a photosensitising agent and light irradiation. Conjugation to biological molecules that specifically target cancer has been shown to increase photosensitiser (PS) selectivity and decrease damage to surrounding normal tissue. The aim of this study was to target tumour cells with a PE-specific PS therefore duramycin was conjugated to a porphyrin based PS which was achieved via direct reaction with the ε-amino group on the lysine residue near duramycin's N-terminal. The compound was subsequently purified using RP-HPLC and confirmed using mass spectrometry. Binding of the conjugate to ovarian and pancreatic cancer cell lines was assessed by flow cytometry. Light irradiation with a light fluence of 7.5J/cm(2) was delivered to conjugate treated cancer cells and cell proliferation analysed by MTT assay. The conjugate detected PE on all 4 cancer cell lines in a concentration dependent manner and conjugate plus irradiation effectively reduced cell proliferation at concentrations ≥0.5µM, dependent on cancer cell line. Reduction in cell proliferation by the irradiated conjugate was enhanced over unconjugated duramycin in A2780, AsPC-1 and SK-OV-3 (p<0.05). In this study we have shown that a duramycin-porphyrin conjugate retained good binding affinity for its target and, following irradiation, reduced cell proliferation of pancreatic and ovarian cancer cell lines.


Bacteriocins/chemistry , Bacteriocins/pharmacology , Peptides/chemistry , Peptides/pharmacology , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Porphyrins/chemistry , Bacteriocins/metabolism , Binding, Competitive , Cell Line, Tumor , Cell Size/drug effects , Cell Size/radiation effects , Humans , Peptides/metabolism , Phosphatidylethanolamines/metabolism , Photosensitizing Agents/metabolism
8.
Cancer Med ; 5(1): 61-73, 2016 Jan.
Article En | MEDLINE | ID: mdl-26590118

In comparison to more differentiated cells, prostate cancer stem-like cells are radioresistant, which could explain radio-recurrent prostate cancer. Improvement of radiotherapeutic efficacy may therefore require combination therapy. We have investigated the consequences of treating primary prostate epithelial cells with gamma irradiation and photodynamic therapy (PDT), both of which act through production of reactive oxygen species (ROS). Primary prostate epithelial cells were cultured from patient samples of benign prostatic hyperplasia and prostate cancer prior to treatment with PDT or gamma irradiation. Cell viability was measured using MTT and alamar blue assay, and cell recovery by colony-forming assays. Immunofluorescence of gamma-H2AX foci was used to quantify DNA damage, and autophagy and apoptosis were assessed using Western blots. Necrosis and senescence were measured by propidium iodide staining and beta-galactosidase staining, respectively. Both PDT and gamma irradiation reduced the colony-forming ability of primary prostate epithelial cells. PDT reduced the viability of all types of cells in the cultures, including stem-like cells and more differentiated cells. PDT induced necrosis and autophagy, whereas gamma irradiation induced senescence, but neither treatment induced apoptosis. PDT and gamma irradiation therefore inhibit cell growth by different mechanisms. We suggest these treatments would be suitable for use in combination as sequential treatments against prostate cancer.


Apoptosis/radiation effects , Autophagy/radiation effects , Cellular Senescence/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Gamma Rays , Prostate/cytology , Apoptosis/drug effects , Autophagy/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Cellular Senescence/drug effects , DNA Damage/drug effects , DNA Damage/radiation effects , Gamma Rays/adverse effects , Humans , Male , Oxidative Stress/genetics , Photochemotherapy , Photosensitizing Agents/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Reactive Oxygen Species/metabolism , Stem Cells
9.
Mol Pharm ; 12(12): 4414-23, 2015 Dec 07.
Article En | MEDLINE | ID: mdl-26559906

Synthesis of the first water-soluble porphyrin radiolabeled with fluorine-18 is described: a new molecular theranostic agent which integrates the therapeutic selectivity of photodynamic therapy (PDT) with the imaging efficacy of positron emission tomography (PET). Generation of the theranostic was carried out through the conjugation of a cationic water-soluble porphyrin bearing an azide functionality to a fluorine-18 radiolabeled prosthetic bearing an alkyne functionality through click conjugation, with excellent yields obtained in both cold and hot synthesis. Biological evaluation of the synthesized structures shows the first example of an (18)F-radiolabeled porphyrin retaining photocytotoxicity following radiolabeling and demonstrable conjugate uptake and potential application as a radiotracer in vivo. The promising results gained from biological evaluation demonstrate the potential of this structure as a clinically relevant theranostic agent, offering exciting possibilities for the simultaneous imaging and photodynamic treatment of tumors.


Fluorine Radioisotopes/chemistry , Porphyrins/chemistry , Radiopharmaceuticals/chemistry , Water/chemistry , Photochemotherapy/methods , Positron-Emission Tomography/methods , Theranostic Nanomedicine/methods
10.
Chem Commun (Camb) ; 51(83): 15304-7, 2015 Oct 25.
Article En | MEDLINE | ID: mdl-26340593

Herein we present a significant step towards next-generation antibody-based photodynamic therapeutics. Site-selective modification of a clinically relevant monoclonal antibody, with a serum-stable linker bearing a strained alkyne, allows for the controlled Cu-free "click" assembly of an in vitro active antibody-based PDT agent using a water soluble azide porpyhrin.


Antibodies, Monoclonal/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology , Alkynes/chemical synthesis , Alkynes/chemistry , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Porphyrins/chemical synthesis , Porphyrins/chemistry , Structure-Activity Relationship
11.
Dalton Trans ; 44(11): 4925-32, 2015 Mar 21.
Article En | MEDLINE | ID: mdl-25437884

The development of novel theranostic agents is an important step in the pathway towards personalised medicine, with the combination of diagnostic and therapeutic modalities into a single treatment agent naturally lending itself to the optimisation and personalisation of treatment. In pursuit of the goal of a molecular theranostic suitable for use as a PET radiotracer and a photosensitiser for PDT, a novel radiolabelled peptide-porphyrin conjugate targeting the α6ß1-integrin has been developed. (69/71)Ga and (68)Ga labelling of an azide-functionalised porphyrin has been carried out in excellent yields, with subsequent bioconjugation to an alkyne-functionalised peptide demonstrated. α6ß1-integrin expression of two cell lines has been evaluated by flow cytometry, and therapeutic potential of the conjugate demonstrated. Evaluation of the phototoxicity of the porphyrin-peptide theranostic conjugate in comparison to an untargeted control porphyrin in vitro, demonstrated significantly enhanced activity for a cell line with higher α6ß1-integrin expression when compared with a cell line exhibiting lower α6ß1-integrin expression.


Metalloporphyrins/chemistry , Peptides/therapeutic use , Photochemotherapy , Photosensitizing Agents/therapeutic use , Positron-Emission Tomography , Alkynes/chemistry , Amino Acid Sequence , Azides/chemistry , Catalysis , Copper/chemistry , Cycloaddition Reaction , Gallium Radioisotopes/therapeutic use , HeLa Cells , Humans , Kinetics , Peptides/chemical synthesis , Peptides/chemistry , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Temperature
12.
Bioconjug Chem ; 25(3): 611-7, 2014 Mar 19.
Article En | MEDLINE | ID: mdl-24564170

The rapidly increasing interest in the synthesis of antibody-drug conjugates as powerful targeted anticancer agents demonstrates the growing appreciation of the power of antibodies and antibody fragments as highly selective targeting moieties. This targeting ability is of particular interest in the area of photodynamic therapy, as the applicability of current clinical photosensitizers is limited by their relatively poor accumulation in target tissue in comparison to healthy tissue. Although synthesis of porphyrin-antibody conjugates has been previously demonstrated, existing work in this area has been hindered by the limitations of conventional antibody conjugation methods. This work describes the attachment of azide-functionalized, water-soluble porphyrins to a tratuzumab Fab fragment via a novel conjugation methodology. This method allows for the synthesis of a homogeneous product without the loss of structural stability associated with conventional methods of disulfide modification. Biological evaluation of the synthesized conjugates demonstrates excellent selectivity for a HER2 positive cell line over the control, with no dark toxicity observed in either case.


Antibodies, Monoclonal, Humanized/chemistry , Immunoglobulin Fab Fragments/chemistry , Photosensitizing Agents/chemistry , Porphyrins/chemistry , Receptor, ErbB-2/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Disulfides/chemistry , Humans , Immunoglobulin Fab Fragments/pharmacology , Molecular Structure , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship , Trastuzumab , Tumor Cells, Cultured
13.
Org Biomol Chem ; 12(8): 1203-6, 2014 Feb 28.
Article En | MEDLINE | ID: mdl-24430855

Fully deprotected alkynyl-functionalised mono- and oligosaccharides undergo CuAAC-based conjugation with water-soluble porphyrin azides in aqueous environments. The mild reaction conditions are fully compatible with the presence of labile glycosidic bonds. This approach provides an ideal strategy to conjugate tetrapyrroles to complex carbohydrates.


Azides/chemistry , Carbohydrates/chemistry , Polysaccharides/chemistry , Porphyrins/chemistry , Solubility , Water/chemistry
...