Supporting Information. Self-Folded Redox/Acid Dual-Responsive Nanocarriers for. Anticancer Drug Delivery



Similar documents
Supplementary Information for

A prochelator with a modular masking group featuring hydrogen peroxide activation with concurrent fluorescent reporting

lung cancer targeted photodynamic therapy and imaging

Supplemental data. A simple and effective cleavable linker for chemical proteomics applications

Human serum albumin (HSA) nanoparticles stabilized with. intermolecular disulfide bonds. Supporting Information

Experimental procedures. Solid phase peptide synthesis (SPPS)

Supporting Information

Solid-phase Synthesis of Homodimeric Peptides: Preparation of Covalently-linked Dimers of Amyloid-beta Peptide

Supplemental Material: peptide synthesis

Supporting Information

Peptide Synthesis Zheng Miao* and Zhen Cheng

Classic Immunoprecipitation

Supporting information. Cyclic peptide-polymer conjugates: grafting to VS grafting from

12.1 Hz, 1H), (m, 11H), (m, 6H), 7.54 (d, J = 8.4 Hz, 2H), (s, 1H), (bs, 1H)

Measuring Cell Viability/Cytotoxicity: Cell Counting Kit-F

A Ratiometric NMR ph Sensing Strategy Based on Slow- Proton-Exchange (SPE) Mechanism

ab Protein Sumoylation Assay Ultra Kit

Chromatin Immunoprecipitation (ChIP)

Supporting Information

How To Make A Tri Reagent

TECHNICAL BULLETIN. FluoroTag FITC Conjugation Kit. Product Number FITC1 Storage Temperature 2 8 C

Western Blot Analysis with Cell Samples Grown in Channel-µ-Slides

SmartFlare RNA Detection Probes: Principles, protocols and troubleshooting

GASTRIC ORGANOID CULTURE PROTOCOL

Supporting Information

Covalent Conjugation to Cytodiagnostics Carboxylated Gold Nanoparticles Tech Note #105

Structure-Based Design of Covalent Siah Inhibitors

CypExpress 3A4 Catalyzed Conversion of Testosterone (TE) to 6β- Hydroxytestosterone (HT)

TECHNICAL REPORT STUDY CHEMICAL PROPERTIES OF RATTAN SHOOT FROM PLANTATION IN THAILAND

MagExtractor -Genome-

Real-time monitoring of rolling circle amplification using aggregation-induced emission: applications for biological detection

Plant Genomic DNA Extraction using CTAB

Guide to Reverse Phase SpinColumns Chromatography for Sample Prep

Novel Method for Solid Phase Peptide Synthesis Using Microwave Energy

Peptide Antibody Production

Creatine Kinase Activity Assay Kit (Colorimetric)

EdU Flow Cytometry Kit. User Manual

Western Blotting: Mini-gels

Peptide synthesis, radiolabelling and radiochemical analysis

Running protein gels and detection of proteins

Evaporation-based Microfluidic Production of. Oil-free Cell-Containing Hydrogel Particles

PROTOCOL. Immunocytochemistry (ICC) MATERIALS AND EQUIPMENT REQUIRED

1. COUPLING REAGENTS : Structure and acronyms

Biology 309 Lab Notebook

Materials and Methods. Protocol for Fmoc- based solid- phase peptide synthesis

Genomic DNA Extraction Kit INSTRUCTION MANUAL

Transfection reagent for visualizing lipofection with DNA. For ordering information, MSDS, publications and application notes see

Marmara Üniversitesi Fen-Edebiyat Fakültesi Kimya Bölümü / Biyokimya Anabilim Dalı PURIFICATION AND CHARACTERIZATION OF PROTEINS

APPLICATIONS MANUAL. Ifosfamide in blood serum Ingredients in blood serum Organophosphorus pesticides in tea leaf... 5

Labelling of peptides with CyDye fluors for fluorescent applications on the LEADseeker Homogeneous Imaging System

RayBio Creatine Kinase (CK) Activity Colorimetric Assay Kit

Small μmol Scale Synthesis of a Labeled Antimicrobial Peptide using Biotage

Fighting the Battles: Conducting a Clinical Assay

Thermo Scientific HyperSep Solid Phase Extraction Method Development Guide

oxidize 4-Cholesten-3-one

Purification of reaction mixtures using flash chromatography.

Enzymes: Amylase Activity in Starch-degrading Soil Isolates

Instructions. Torpedo sirna. Material. Important Guidelines. Specifications. Quality Control

PRODUCT INFORMATION...

RPCI 004 v.002 Staining Procedure For all Directly Conjugated Reagents (Whole Blood Method)

Oasis HLB Cartridges and 96-Well Plates

Alkoxycarbonylation of Ethylene with Cellulose in Ionic Liquids

protocol handbook 3D cell culture mimsys G hydrogel

BD PuraMatrix Peptide Hydrogel

EXPERIMENT 5: DIPEPTIDE RESEARCH PROJECT

SPE and HPLC. Dr Iva Chianella Lecturer in Analytical Chemistry Cranfield Health +44 (0)

LUMEFANTRINE Draft proposal for The International Pharmacopoeia (October 2006)

ph-induced outward Movement of Star Centers within Coumarin-Centered Star-Block Polymer Micelles Supporting Information

INSTRUCTION Probemaker

Creatine Kinase Activity Colorimetric Assay Kit ABE assays; Store at -20 C

Supporting information for Journal of Materials Chemistry

ab FITC Conjugation Kit Protocol

NimbleGen DNA Methylation Microarrays and Services

Laboratory procedures in polymer chemistry

Working with Hazardous Chemicals

Z-Stacking and Z-Projection using a Scaffold-based 3D Cell Culture Model

ReadyPrep Protein Extraction Kit (Soluble/Insoluble) Instruction Manual. Catalog #

Lab 10: Bacterial Transformation, part 2, DNA plasmid preps, Determining DNA Concentration and Purity

Rat Creatine Kinase MB Isoenzyme (CKMB) ELISA

LIVE/DEAD Fixable Dead Cell Stain Kits

Supplementary Information

Acid-Base Extraction.

Standard Operating Procedure (SOP)

ArC Amine Reactive Compensation Bead Kit

TransIT Transfection Reagent

Supplementary Information. Primary amino acid lithium salt as a catalyst for asymmetric Michael addition of isobutyraldehyde with β-nitroalkenes.

Studying Photosynthetic Electron Transport through the Hill Reaction

An In-Gel Digestion Protocol

Glycolysis Cell-Based Assay Kit

BUFFERS and MEDIAS Coomassie Blue Staining Solution Coomassie blue Destaining Solution DMEM Normal Cell Culture Media

HighPure Maxi Plasmid Kit

HiPer Ion Exchange Chromatography Teaching Kit

Experiment 3: Extraction: Separation of an Acidic, a Basic and a Neutral Substance

PCR and Sequencing Reaction Clean-Up Kit (Magnetic Bead System) 50 preps Product #60200

and its application in the synthesis of Nilotinib intermediate

PROTOCOL. Immunostaining for Flow Cytometry. Background. Materials and equipment required.

RiboZol RNA Extraction Reagents

TRI Reagent Solution. A. Product Description. RNA / DNA / Protein Isolation Reagent Part Number AM ml

Protein extraction from Tissues and Cultured Cells using Bioruptor Standard & Plus

Data Sheet. PD-L1:B7-1[Biotinylated] Inhibitor Screening Assay Kit Catalog # Size: 96 reactions

Transcription:

Electronic Supplementary Material (ESI) for ChemComm. This journal is The Royal Society of Chemistry 2014 Supporting Information Self-Folded Redox/Acid Dual-Responsive Nanocarriers for Anticancer Drug Delivery Yue Lu ab, Ran Mo ab, Wanyi Tai ab, Wujin Sun ab, Dennis B. Pacardo ab, Chenggen Qian abc, Qundong Shen c, Frances S. Ligler a and Zhen Gu ab * a Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA. E-mail: zgu@email.unc.edu; zgu3@ncsu.edu. b Center for Nanotechnology in Drug Delivery and Division of Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA. c Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing, China. 1

Experimental Section Materials All chemicals were obtained from commercial sources and used without further purification. Tris(2-aminoethyl)amine, di-t-butyl dicarbonate (Boc 2 O), trifluoroacetic acid (TFA), N,O-bis(trimethylsilyl) acetamide (BSA) and triphosgene were purchased from Sigma Aldrich. N,N'-diisopropylcarbodiimide (DIPC), triethylamine (Et 3 N) and 4-dimethylaminopyridine (DMAP) were obtained from Acros Organics. NHS- PEG 5000 -NHS, NHS-PEG-SS-PEG-NHS and Folic acid-peg-ma were purchased from Nanocs Inc. 2-Ethoxypropene was purchased from. Doxorubicin (DOX) was from Alfa Aesar. All the organic solvents for synthesis and analysis were ordered from Fisher Scientific Inc. and used as received. Methods Synthesis of graft copolymer. Synthesis of monomer I. N, N-bis(2-aminoethyl)-N-[2-(tert-butylcarbamoyl) ethylamine] (monomer I) was synthesized as reported. Boc 2 O (1 g, 4.6 mmol, chloroform solution) was added dropwise into tris(2-aminoethyl)amine (6.7 g, 46 mmol) dissolved in 300 ml of chloroform at 0 o C, under stirring. The reaction was allowed to warm up to room temperature and stirred overnight. The reaction was quenched by 15 ml of DD water. The organic phase was separated after being stirred for 5 min. The aqueous phase was re-extracted with 30 ml chloroform. The combined organic phase was first dried with Na 2 SO 4, then concentrated under vacuum to yield crude compound, which was further purified with silica gel chromatography using CHCl 3 /MeOH/concentrated aqueous NH 4 OH (v/v/v, 10/5/1) as eluent. The purified monomer I was given as viscous oil. Synthesis of monomer IV. AcO-L-Serine (500 mg, 3.4 mmol) was dissolved in 20mL tetrahydrofuran (THF), followed by the addition of triphosgene (392 mg, 1.7 mmol). The mixture was allowed to react under 48 o C for 2 h. The resulted product solution was first purified with silica gel chromatography using petroleum ether/acoet (v/v, 2/1 for the 1 st elution, 1/1 for the 2 nd elution) as eluent. The product solution was then rotary evaporated to give light yellow oil. Synthesis of multiblock copolymer by condensation polymerization. Multiblock copolymerwas synthesized via one-pot polymerization. Briefly, monomer I, NHS- PEG 5000 -NHS (monomer II) and NHS-PEG-SS-PEG-NHS (monomer III) were predried in CaCl 2 desiccator for two days before synthesis. NHS-PEG 5000 -NHS (200 mg, 40 μmol) and NHS-PEG-SS-PEG-NHS (14.8 mg, 13 μmol) were dissolved in 2 ml DMF, followed by the addition of 132 μl monomer I (13.8 mg, 50 μmol) solution in DMF under N 2 atmosphere. The reaction was initiated by adding 22.3 µl of Et 3 N. Two days later, the reaction mixture was poured into 50 ml diethyl ether. The precipitate was washed twice with diethyl ether, dried in vacuum and yield as white powder. 2

TMS activation of linear multi-block copolymer. To remove the protecting group Boc, The above resulting copolymer was dissolved in 10 ml of dichloromethane (DCM) and TFA mixture (v/v, 1/1). The scavenger, phenol (70 mg), was also added into the mixture to minimize the side reaction. The reaction mixture was stirred at room temperature for 1 hour. The organic solvent was evaporated in vacuum and the residue was washed with diethyl ether. After removing the remaining organic solvent in vacuum, the residue was dissolved in water and dialysis against DD water with cellulose tubing (MWCO: 12 kda). The resulting solution was lyophilized and dried in CaCl 2 desiccator. The amine groups on the multiblock copolymer were activated with TMS as initiator for graft polymerization. The dry polymer (100 mg) was mixed with 5 ml of anhydrous benzene. After polymer was completely dissolved, 2 ml of N,O-bis(trimethylsilyl) acetamide (BSA) was added. The reaction was stirred at room temperature for 24 h. Anhydrous hexane (10 ml) was added into the reaction mixture to precipitate the product. The white precipitate was washed three times with anhydrous hexane and dried under vacuum to yeild TMS-activated multi-block copolymer. Graft polymerization. The Ring-Opening Polymerization of the side chain was initiated by N-TMS Amines. Briefly, monomer IV (127 mg) and TMS-activated multi-block copolymer (97.3 mg) was suspended in 5 ml of dry chloroform. The yellow suspension gradually turned clear after adding 20 µl of Et 3 N. The reaction gradually became turbid over time, indicating the successful polymerization. Forty eight hours later, the reaction mixture was poured into 30 ml of diethyl ether to precipitate the product. After washing three times with diethyl ether, the precipitate was dried under vacuum to give graft copolymer. The yielded graft copolymer was dissolved in water with the addition of LiOH. The hydrolytic was then dried under vacuum. Modification of side chains. 2-Ethoxypropene was added to the DMSO solution of graft copolymer with the existence of PPTS under magenitic stirring. After reacting for 1 h, the reaction process was quenched by TEA. Preparation and characterization of DOX/RAD-NCs. To obtain self-folded RAD-NCs, 1 ml of the graft copolymer solution (dissolved in DMSO) was drop-wisely injected (2mL h -1 ) into 4 ml of DD water under vigorously magnetic stirring. The RAD-NCs solution was then transferred into centrifuge tube and sonicated for 30 cycles (1 s each with a duty cycle of 20%). The homogenous RAD-NCs solution was dialyzed against DD water for 48 hours at room temperature in order to completely get rid of organic solvent DMSO. The DD water was frequently changed during the dialysis process. The folded RAD-NCs were stored at 4 o C for further characterization. In order to encapsulate DOX into RAD-NCs, DOX HCl was added into DMSO together with TEA under stirring. 1 ml of graft copolymer solution was then added into the above reaction mixture. The resulting homogenous solution was injected slowly into DD water as described above. 3

In vitro drug release. The release profiles of DOX were assayed using dialysis method in PBS buffer as reported. [50] 400 μl DOX/RAD-NCs solution with 14 mg of DOX was added into a dialysis tube (10 K MWCO) (Slide-A-Lyzer, Thermo Scientific) against 14 ml of PBS buffer solution with different ph (7.4 and 5.0) and different concentrations of GSH (0, 1 mm and 10 mm). The dialysis tube was immerged in 37 C water bath. At predetermined time intervals, the total buffer solution was withdrawn, followed by replacing with 14 ml of fresh buffer solution with the same ph and GSH concentration. The fluorescence intensity of released DOX was measured at 596nm with an excitation wavelength of 480 nm by microplate reader (Infinite M200 PRO, Tecan). Insertion of target ligand. GSH (0.5 mm) was added into 2 ml DOX/RAD-NCs solution under magnetic stirring for 5 min, dialyzed against DD water with cellulose tubing (MWCO: 12 kda), followed by the adding of folic acid-peg-maleimide (1 mg, 0.5 μmol). After stirred for 1 h, the reaction mixture was dialysed against DD water with cellulose tubing (MWCO: 12 kda). Cell culture. HeLa cells were obtained from Tissue Culture Facility of UNC Lineberger Comprehensive Cancer Center and cultured in DMEM with 10% (v:v) fetal bovine serum (FBS), 100U ml -1 penicillin and 100 mg ml -1 streptomycin in an incubator (Thermo Scientific) at 37 under an atmosphere of 5% CO 2 and 90% relative humidity. The cells were sub-cultivated approximately every 3 days at 80% confluence using 0.25% (w: v) trypsin at a split ratio of 1:5. Cellular uptake mechanism. HeLa cells (1 10 5 cells per well) were seeded in 6-well plates. After culture for 48 h, the cells were pre-incubated with several specific inhibitors for various kinds of endocytosis [inhibitor of clathrin-mediated endocytosis: sucrose (SUC, 450 mm) and chlorpromazine (CPZ, 10 μm); inhibitor of caveolin-mediated endocytosis: nystatin (NYS, 25 μg ml -1 ); inhibitor of macropinocytosis: amiloride (AMI, 1 mm); inhibitor of lipid raft: methyl-β-cyclodextrin (MCD, 3 mm)] for 1 h at 37 C, respectively. Afterward, the cells were incubated with DOX/RAD-NCs at a DOX concentration of 1 mg/l in the presence of inibitors for additional 2 h. After washing the cells by PBS twice, the fluorescence intensity of DOX in the cells and the cell proteins were measured, respectively. Intracellular trafficking. 4

HeLa cells (1 10 5 cells per well) were seeded in a confocal microscopy dish (MatTek). After culture for 24 h, the cells were incubated with DOX/RAD-NCs and DOX/FA-RAD-NCs (2 mm Dox concentration) at 37 C for 1 h and 4 h, respectively, and then washed by PBS twice. Subsequently, the cells were stained by LyosTracker Green (50 nm) (Life Technologies) at 37 C for 30 min and Hoechst 33342 (1 mg ml - 1 ) (Life Technologies) at 37 C for 10 min. Finally, the cells were washed by PBS twice and immediately observed using CLSM (LSM 710, Zeiss). In vitro cytotoxicity. HeLa cells (1 10 4 cells per well) were seeded in 96-well plates. After 24 h culture, the cells were exposed to the PBS buffer, free DOX solution, DOX/RAD-NCs and DOX/FA-RAD-NCs with different concentrations of DOX for 24 h and 48 h, respectively. 20 μl MTT solution (5 mg ml -1 ) was then added to each well. After 4 h of incubation, the medium was removed, and 150 ml DMSO was added to each well. The absorbance was measured at a test wavelength of 570 nm and a reference wavelength of 630 nm by microplate reader (Infinite M200 PRO, Tecan). 5

Figure S1. Synthesis route of graft copolymer. Monomer I, monomer II and monomer III were firstly condensed into a linear multiblock copolymer by NHS mediated amidation. After liberating the third amino group of monomer I, the side chains were gradually elongated on the main chain via a TMS controlled ROP of monomer IV. The side chains were then modified with 2-ethoxypropene into the target graft copolymer capable of self-folding. 6

Figure S2. Synthesis route of AcO-L-Serine-NCA (monomer IV). 7

Figure S3. Representative TEM images of DOX/RAD-NCs. Scale bar 200 nm. 8

Figure S4. Representative TEM images of A) Folic-acid conjugated RAD-NCs. B) DOX/ FA-RAD-NCs. C) FA-RAD-NCs treated with GSH and acid (ph 5.0). Scale bars 100 nm. 9

Figure S5. Relative uptake efficiency of DOX/RAD-NCs on HeLa cells in the presence of various endocytosis inhibitors. Error bars indicate s.d. (n = 3). *P < 0.05, **P < 0.01 compared with the control group (two-tailed Student s t-test). Inhibitor of clathrin-mediated endocytosis: sucrose (SUC) and chlorpromazine (CPZ); inhibitor of caveolin-mediated endocytosis: nystatin (NYS); inhibitor of macropinocytosis: amiloride (AMI); inhibitor of lipid raft: methyl-β-cyclodextrin (MCD). Compared with the cellular uptake of DOX/RAD-NCs without inhibitors as a control, the significant decrease in uptake of DOX/RAD-NCs with inhibitors confirmed the corresponding endocytosis pathways of the nanocarriers. 10

Figure S6. Intracellular trafficking of DOX/RAD-NCs (A) and DOX/FA-RAD-NCs (B) on HeLa cell line observed by CLSM (large cell numbers). The late endosomes and lysosomes were stained by LysoTracker Green, and the nuclei were stained by Hoechst 33342. Scale bar: 20 μm. 11

Figure S7. Cytotoxicity study of empty RAD-NCs and FA-RAD-NCs after 48 hour culture with HeLa cells. The horizontal ordinate indicates the concentration of RAD- NCs and FA-RAD-NCs. Error bars indicate s.d. (n=4). 12