Enzymatic Biodegradation of Hydrogels for Protein Delivery Targeted

Feb 12, 2015 - Accounts of Chemical Research 2017 50 (2), 170-178 ... Synthesis, characterization and application in multiple responses hydrogel. Xuze...
0 downloads 0 Views 3MB Size
Subscriber access provided by University of Cincinnati Libraries

Article

Enzymatic biodegradation of hydrogels for protein delivery targeted to the small intestine Jennifer M Knipe, Frances M Chen, and Nicholas A Peppas Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/bm501871a • Publication Date (Web): 12 Feb 2015 Downloaded from http://pubs.acs.org on February 18, 2015

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 41

Biomacromolecules

1 2 3 4 6

5

Enzymatic biodegradation of hydrogels for protein 8

7 9 1

10

delivery targeted to the small intestine 12 13 14 16

15

Jennifer M. Knipe a, Frances Chen a, Nicholas A. Peppas a,b,c* 17 18 20

19

a Department of Chemical Engineering, C0400, The University of Texas at Austin, Austin, TX 2

21

78712, USA 23 24 25

b Department of Biomedical Engineering, C0800, The University of Texas at Austin, Austin, TX 26 28

27

78712, USA 29 31

30

c College of Pharmacy, C0400, The University of Texas at Austin, Austin, TX 78712, USA 32 3 34 35 37

36

KEYWORDS hydrogel, biodegradation, trypsin, peptide, intestinal delivery 38 39 41

40

Multi-responsive poly(methacrylic acid-co-N-vinyl pyrrolidone) hydrogels were synthesized 43

42

with biodegradable, oligopeptide crosslinks. The oligopeptide crosslinks were incorporated using 4 45

EDC-NHS zero-length links between the carboxylic acid groups of the polymer and free primary 46 48

47

amines on the peptide. The reaction of the peptide was confirmed by primary amine assay and IR 50

49

spectroscopy. The microgels exhibited pH-responsive swelling as well as enzyme-catalyzed 51 53

52

degradation targeted by trypsin present in the small intestine, as demonstrated upon incubation 5

54

with gastrointestinal fluids from rats. Relative turbidity was used to evaluate enzyme-catalyzed 56 57 58 59 60 ACS Paragon Plus Environment

1

Biomacromolecules

Page 2 of 41

1 3

2

degradation as a function of time, and initial trypsin concentration controlled both the 5

4

degradation mechanism as well as the extent of degradation. Trypsin activity was effectively 6 8

7

extinguished by incubation at 70°C, and both the microgels and degradation products posed no 10

9

cytotoxic effect to two different cell lines. The microgels demonstrated pH-dependent loading of 1 12

the protein insulin for oral delivery to the small intestine. 13 14 15 16 17 18 19 20 21 2 23 24 25 26 27 28 29 30 31 32 3 34 35 36 37 38 39 40 41 42 43 4 45 46 47 48 49 50 51 52 53 54 5 56 57 58 59 60 ACS Paragon Plus Environment

2

Page 3 of 41

Biomacromolecules

1 3

2

Introduction 4 6

5

Environmentally-responsive hydrogels, or hydrophilic, crosslinked polymer networks that 7 9

8

undergo physicochemical changes in response to one or more environmental stimuli, offer the 1

10

specificity of highly tunable materials combined with excellent biocompatibility 1-4. As the next 12 13

generation of biomaterials, these “intelligent” networks are able to respond to or mimic 14 15

biological environments and processes such as vascularization 5, 6, tumor physiology 7, 8, 18

17

16

endosomal compartments 9-11, or the extracellular matrix 12, 13. This capability could be 19 20

instrumental in achieving various biomedical advances, including tissue regeneration and 21 23

2

controlled delivery of biological therapeutics 14, 15. 24 26

25

Hydrogels with pH-responsive behavior are among the most widely utilized “intelligent” 27 28

hydrogel systems for drug delivery applications 16. Polyanionic hydrogels such as 31

30

29

poly(methacrylic acid) (PMAA) exhibit complexation via hydrogen bonding at low pH 32 3

conditions, such as that of gastric fluid, and undergo increased swelling due to ionization of the 34 35

carboxylic groups at neutral pH conditions, such as that of the intestine 17. Thus, PMAA 38

37

36

copolymers have been widely utilized as oral drug delivery carriers or coatings for their ability 39 40

to protect a loaded therapeutic from denaturation and proteolytic degradation as it travels through 41 43

42

gastric conditions yet swell and release the therapeutic at the site of absorption in the small 45

4

intestine 18-23. 46 47 49

48

Biodegradation is another possible environmental response of hydrogels designed as drug 51

50

delivery applications 14, 24. Polymers that degrade by hydrolysis, such as polyanhydrides 25, 26, 53

52

poly(orthoesters) 2, 27, poly(caprolactone), and poly(lactic acid) and poly(glycolic acid) 28, 29 are 54 56

5

widely used for drug delivery. However, polymers that undergo enzyme-catalyzed degradation 57 58 59 60 ACS Paragon Plus Environment

3

Biomacromolecules

Page 4 of 41

1 3

2

are becoming attractive as site-specific delivery vehicles due to the localized concentration of 5

4

enzymes throughout the body as well as the specificity of enzymatic attack 30, 31. A relatively 6 8

7

small number of researchers have investigated the use of hydrogels with enzyme-degradable 10

9

peptide components for the purpose of drug delivery to the small intestine 30, 32, 33, where 12

1

enzymes such as trypsin, chymotrypsin, and cathepsin-β are prevalent 34. In particular, peptide 13 15

14

crosslinks are an appealing route to achieve enzyme-specific degradation of the network and 17

16

subsequent drug delivery 30, 32, 33, 35, 36. For example, oligopeptide crosslinks of different lengths 18 19

were incorporated into poly(N-(2-hydroxypropyl)- methacrylamide) (PHPMA) hydrogels, and 20 2

21

the release of macromolecules from the gels upon exposure to chymotrypsin or liver enzymes 24

23

was determined to be a function of both peptide length and structure 33. Wanakule et al. 35 25 27

26

showed the rapid drug release from peptide-crosslinked PLGA microspheres triggered by trypsin 29

28

reached a plateau after only hours, compared to hydrolysis and diffusion mediated release that 31

30

occurred over days. Both the control and timescale of the peptide degradation are attractive 32 34

3

characteristics for drug delivery, particularly via an oral route. 35 37

36

Here, poly(methacrylic acid-co-N-vinylpyrrolidone) (P(MAA-co-NVP)) polymer chains 38 39

were used to impart hydrophilic and pH-responsive behavior that either inhibited or allowed 40 42

41

diffusion of enzymes into the polymer network due to pH-responsive complexation. The polymer 4

43

chains were crosslinked by a facile bioconjugation reaction with an oligopeptide rich in arginine 45 46

and lysine groups targeted for degradation specifically by the enzyme trypsin. Synthesis, 47 49

48

degradation, cytocompatibility, and therapeutic loading and release using the pH-responsive 51

50

P(MAA-co-NVP) crosslinked by biodegradable peptide are detailed herein. 52 53 5

54

Experimental 56 57 58 59 60 ACS Paragon Plus Environment

4

Page 5 of 41

Biomacromolecules

1 3

2

Materials 4 6

5

Methacrylic acid (MAA), N-vinyl-2-pyrrolidone (NVP), Irgacure 184® (1-hydroxy7 9

8

cyclohexyl-phenylketone), 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride 1

10

(EDC), purified pepsin from porcine gastric mucosa (≥2500 U/mg) and pancreatin from porcine 12 13

pancreas (4x USP specifications), trypsin-EDTA solution (1X) and Nα-benzoyl-L-arginine ethyl 14 16

15

ester hydrochloride (BAEE) trypsin substrate, and recombinant human insulin (≥27.5 IU/mg) 18

17

were purchased from Sigma-Aldrich (St. Louis, MO). N-hydroxysuccinimide (NHS) was 19 20

purchased from Pierce Biotechnology, Inc. (Rockford, IL). The custom sequence oligopeptide 21 23

2

GRRRGK was synthesized by CHI Scientific (Maynard, MA). Fluorescamine was purchased 25

24

from Acros Organics (Geel, Belgium). All reagents were used as received. All other solvents and 26 28

27

buffers were purchased from Fisher Scientific (Waltham, MA). 29 31

30

Methods 32 3 34

Synthesis and Purification 35 36 38

37

P(MAA-co-NVP) linear polymer was synthesized by photoinitiated, free-radical 40

39

polymerization. MAA and NVP were added at a 1:1 molar ratio to a 1:1 (w/w) deionized water 41 42

and ethanol solution to yield a 1:3 (w/w) total monomer to solvent ratio. Photoinitiator Irgacure 43 45

4

184® was added at 1 wt% with respect to total monomer weight. The mixture was homogenized 47

46

by sonication then the flask was sealed with a rubber septum. The solution was purged with 48 50

49

nitrogen for 20 minutes, then the polymerization was initiated with a Dymax BlueWave® 200 52

51

UV point source (Dymax, Torrington, CT) at 100mW/cm2 intensity and allowed to polymerize 54

53

for 30 minutes while stirring. 5 56 57 58 59 60 ACS Paragon Plus Environment

5

Biomacromolecules

Page 6 of 41

1 3

2

Following polymerization, the linear polymer was purified from unreacted monomer by 5

4

addition of 1 N hydrochloric acid (HCl) to precipitate polymer, centrifugation, and resuspension 6 8

7

in deionized water. After three wash cycles, the polymer solution was neutralized, frozen in 10

9

liquid nitrogen, and lyophilized. 1 12 13

To synthesize the peptide-crosslinked hydrogels, linear P(MAA-co-NVP) was dissolved in a 14 16

15

1:1 (v/v) water:ethanol solution at a concentration of 50 mg/ml. EDC was dissolved in ethanol at 18

17

a concentration of 50 mg/ml and NHS was dissolved in ethanol at a concentration of 16 mg/ml. 19 20

The EDC and NHS solutions were added to the polymer solution at a ratio of 6:3:1 21 23

2

polymer:EDC:NHS by weight. The solution was mixed by vortex, then allowed to react for ~3 25

24

min with shaking. The pH was raised to ~8 by the addition of 1 N sodium hydroxide (NaOH), 26 28

27

and then a volume of 100 mg/ml peptide in ethanol solution was added to achieve a 2:1 weight 30

29

ratio of polymer:peptide. The mixture was allowed to react overnight with shaking then purified 32

31

by three wash cycles with water and centrifugation at 10,000 x g for 5 minutes. Following the 3 35

34

washes, the polymer was frozen in liquid nitrogen and lyophilized for at least 24 hours. 36 38

37

After lyophilization, the polymer was milled into a fine power by crushing with mortar and 39 40

pestle. The powder was sifted to the size ranges of 30-75 µm and less than 30 µm by 41 43

42

ultraprecision ASTM sieves (Precision Eforming, Cortland, NY). 4 46

45

Characterization 47 48 50

49

Potentiometric Titration 51 53

52

To determine the MAA content of the linear polymer, a 3.5 mg/ml solution of polymer in 54 5

deionized water was titrated to pH 11.5 using 0.2 N NaOH (standardized with potassium 56 57 58 59 60 ACS Paragon Plus Environment

6

Page 7 of 41

Biomacromolecules

1 3

2

hydrogen phthalate) at 25°C with constant stirring. pH was measured with a Mettler-Toledo 5

4

SevenEasy™ (Columbus, OH) pH probe and was recorded when the pH reached a steady value 6 8

7

(±0.01 pH units in three consecutive measurements over 5 minutes). The equivalence point was 10

9

used in conjunction with a charge balance to determine the amount of MAA present in each 1 12

formulation. 13 14 16

15

Fluorescamine Assay 17 19

18

The fluorescamine solution was prepared fresh before each test by dissolving 3 mg of 20 21

fluorescamine in 10 ml filtered acetone. Supernatant from the EDC-NHS reactions was mixed in 2 24

23

a range of dilutions with phosphate buffered saline (PBS) and the fluorescamine solution with 26

25

agitation. After reacting at room temperature with shaking for 15 min, 200 µl of each sample 27 29

28

was transferred in triplicate to a black 96-well plate and the fluorescence at 360 ex/460 em was 31

30

measured using a Bio-Tek Synergy™ HT multi-mode plate reader(Winooski, VT), 32 3

sensitivity=85. 34 35 37

36

Fourier Transform Infrared Spectroscopy 38 40

39

Fourier transform infrared spectroscopy (FTIR) spectra were obtained using a Thermo Mattson 41 42

Infinity Gold spectrometer (Thermo Fisher Scientific Inc., Waltham, MA). The incubation buffer 43 45

4

of degraded hydrogel samples was exchanged with water using 30,000 MWCO centrifugal filters 47

46

(Millipore, Billerica, MA) over 5 washes. Samples were lyophilized and then pressed in KBr 48 49

(Sigma-Aldrich) disks. For each sample, 512 scans were performed with a resolution of 4 cm-1 50 52

51

and gain of 1.0, and background spectra of a KBr blank disk was subtracted from the sample 54

53

spectra. 5 56 57 58 59 60 ACS Paragon Plus Environment

7

Biomacromolecules

Page 8 of 41

1 3

2

Scanning Electron Microscopy 4 6

5

Scanning electron microscopy (SEM) samples were prepared by dusting carbon tape-covered 7 9

8

aluminum stubs with lyophilized, crushed microgels. The samples were coated with 8-10 nm of 1

10

Pt/Pd coating using a Cressington 208 Benchtop sputter coater (Watford, England). Scanning 12 13

electron microscopy images were obtained using an FEI Quanta 650 FEG scanning electron 14 16

15

microscope (Hillsboro, OR) and a Zeiss Supra 40V scanning electron microscope (Jena, 18

17

Germany). 19 20 2

21

Degradation 23 25

24

Microgels were degraded at various trypsin concentrations in 1X phosphate buffered saline 27

26

solution (pH 7.4), simulated gastric fluid, simulated intestinal fluid, rat gastric fluid or rat 28 30

29

intestinal fluid. Simulated gastric fluid (SGF) and simulated intestinal fluid (SIF) were prepared 32

31

according to USP 29 37. Briefly, the SGF was prepared by dissolving 2 g of sodium chloride and 3 34

3.2 g of purified pepsin from porcine stomach mucosa in ~800 ml deionized water. 7 ml of HCl 35 37

36

was added, followed by enough water to make up to 1 L and the pH adjusted to 1.2. SIF was 39

38

prepared by dissolving 6.8 g monobasic potassium phosphate in 250 ml deionized water, then 77 40 41

ml of 0.2 N NaOH was added while stirring. 500 ml additional water was added then 10 g 42 4

43

pancreatin was mixed into the solution. The pH was adjusted to 6.8 using 0.2 N NaOH or HCl 46

45

then the solution was made up to 1 L with water. 47 48 50

49

Gastrointestinal fluids were harvested from Sprague Dawley juvenile male rats (250-300 g) 52

51

according to a protocol published by Yamagata et al. with some modifications 38. Briefly, after 54

53

sacrificing the rat the stomach was excised and ligated at both ends. A needle was inserted to 5 57

56

inject 5 ml of pH 1.2 HCl-NaCl buffer (same as SGF minus pepsin) and the gastric contents were 58 59 60 ACS Paragon Plus Environment

8

Page 9 of 41

Biomacromolecules

1 3

2

collected in a 50 ml centrifuge tube. Similarly, a ~20 cm section of the upper small intestine was 5

4

cannulated and flushed twice with 10 ml cold PBS (1X, pH 7.4). The fluid was collected as 6 8

7

intestinal fluid in a 50 ml centrifuge tube. Both the harvested fluids were centrifuged at 3,200 x 10

9

g, 4°C, for 15 min to separate solids from the fluids. The supernatants were retained as rat gastric 1 12

fluid and rat intestinal fluid, respectively. Fluids were stored at -20°C until use. 13 14 16

15

Degradation was measured by relative turbidity of the solutions over time, as reported by 18

17

Klinger and Landfester 39. Microgels were suspended in trypsin solutions of varying 19 20

concentration, PBS, SGF, SIF, or rat gastrointestinal fluids at various concentrations. 100 µl of 21 23

2

each sample solution, including pure solutions of PBS, SGF, SIF, or rat gastrointestinal fluids as 25

24

a control, was added to a 96-well plate in triplicate. The absorbance was then measured at 500 26 28

27

nm in 5 minute intervals over 90 minutes using a Bio-Tek Synergy™ HT multi-mode plate 30

29

reader (Winooski, VT). The temperature was controlled at 37°C and the plate underwent shaking 32

31

for 3 seconds before each measurement. 3 34 36

35

Activity of the trypsin following incubation with particles and deactivation methods including 38

37

addition of serum-containing cell culture media or 5 minutes incubation at 60°C, 70°C, or 80°C, 40

39

was evaluated using a trypsin activity assay adapted from the protocol by Yanes et al. 40. Briefly, 41 43

42

degradation supernatant was combined with 1 mg/ml BAEE in PBS at a 1:9 sample:BAEE ratio 45

4

by volume. Immediately after addition of the BAEE, absorbance at 253 nm was measured at the 46 47

minimum interval (typically 40-50 seconds) for 5 minutes using a Bio-Tek Synergy™ HT multi48 50

49

mode plate reader (Winooski, VT). 51 53

52

In vitro Cytotoxicity Study 54 5 56 57 58 59 60 ACS Paragon Plus Environment

9

Biomacromolecules

Page 10 of 41

1 3

2

L929 and RAW 264.7 cell lines were obtained from American Type Culture Collection 5

4

(ATCC, Rockwell, MD). All cell lines were cultured in Dulbecco’s modified Eagle medium 6 8

7

(DMEM) (Mediatech, Herndon, VA) supplemented with 10% heat-inactivated HyClone™ Fetal 10

9

Bovine Serum, USDA Tested (Fisher Scientific), 1% 200 mM L-glutamine solution (Mediatech), 1 12

100 U/ml penicillin, and 100 µg/ml streptomycin (Mediatech). Cytotoxicity studies were 13 15

14

performed using DMEM without phenol red supplemented with 2% heat-inactivated HyClone™ 17

16

Fetal Bovine Serum, USDA Tested (Fisher Scientific), 1% non-essential amino acids 18 19

(Mediatech), 100 U/ml penicillin, and 100 µg/ml streptomycin (Mediatech). Cells were 20 2

21

incubated at 37°C in a 5% CO2 environment. 23 25

24

Cells were seeded at a density of 10,000 cells/well in a 96-well plate and allowed to incubate 26 28

27

for 24 hours prior to the experiment. Microgels were degraded in 1.25 or 0.625 mg/ml trypsin in 30

29

PBS at concentrations ranging from 1.3-6 mg/ml. Degradation took place at 37°C with shaking 32

31

for at least 4 hours. Trypsin was deactivated by addition of 2X volume DMEM without phenol 3 35

34

red containing 2% fetal bovine serum. Cells were incubated with degraded microgels for 8 hours 37

36

at 37°C and 5% CO2. The cytotoxic effect of the microgels was evaluated using a CellTiter 96® 38 39

Aqueous One Solution Cell Proliferation MTS Assay (Promega, Madison, WI). MTS assay was 40 42

41

added to the wells and incubated for 90 minutes at the same conditions before absorbance 4

43

measurements were made at 490 nm using a Bio-Tek Synergy™ HT multi-mode plate reader 45 46

(Winooski, VT). Cytotoxicity is reported as ‘relative cell proliferation’, or normalization of the 47 49

48

assay absorbance values to the average assay absorbance for cells incubated in only culture 51

50

media. 52 53 5

54

Insulin Loading 56 57 58 59 60 ACS Paragon Plus Environment

10

Page 11 of 41

Biomacromolecules

1 3

2

Microgels were loaded by equilibrium partitioning post-synthesis with recombinant human 5

4

insulin. Microgels were incubated at 37°C for 4 hours in a 0.5 mg/mL insulin solution of pH ~5.5 6 8

7

at a ratio of 7:1 microgel:therapeutic by weight. The microgels were collected by centrifugation 10

9

at 10,000 x g for 5 minutes then collapsed by resuspension in 0.5 N HCl. Microgels were 1 12

separated from supernatant by centrifugation at 10,000 xg for 5 minutes. The loaded microgels 13 15

14

were lyophilized and stored at -20°C for further studies. Protein loading was evaluated with a 17

16

MicroBCA assay protein quantification assay (Pierce-Thermo, Rockford, IL). 18 19 20

Results and Discussion 21 23

2

Synthesis, purification and lyophilization of the uncrosslinked P(MAA-co-NVP) yielded 25

24

white, fluffy polymer. Due to the phase transition of MAA from hydrophobic to hydrophilic 26 28

27

above pH ~5, adjusting the pH of the solution to neutral prior to freeze drying facilitated the 30

29

solubilization of the dried polymer into aqueous solution. Potentiometric titration was used to 32

31

determine that the linear polymer was approximately 45 mol% or 39 wt% MAA. 3 35

34

The number average molecular weight of linear chains, Mn, may be calculated from the 37

36

kinetics of free-radical polymerization for each polymer 41. 39

38

  

 =  42

41

40

(1)

/    

Values for the rate constants were found for both methacrylic acid and N-vinylpyrrolidone42, 45

4

43

the initiator efficiency, f, was approximated as 0.75, and the value of a, a constant related to the 46 47

method of termination was 1.33 48

41

. Actual values were input for monomer concentration [M],

50

49

initiator concentration [I], and molecular weight of the repeat unit M0. The number average 52

51

molecular weight of linear chains of each polymer was found to be on the order of 10,000, which 54

53

in in agreement with other values reported 43, thus linear chains of the copolymer are expected to 5 57

56

be on the same order. 58 59 60 ACS Paragon Plus Environment

11

Biomacromolecules

Page 12 of 41

1 3

2

Scheme 1 shows the mechanism of the EDC-NHS crosslinking reaction with the linear 5

4

P(MAA-co-NVP). Upon solubilization of the polymer in the ethanol-water solution, the pH was 6 8

7

adjusted to ~5 to favor the activation of the carboxylic acid groups by EDC and increase the 10

9

stability of the active ester intermediate 44. EDC was added at a molar ratio of 1:2 to the MAA 1 12

groups on the linear chains, and NHS was added at a molar ratio of 1:1.8 to the EDC. Upon 13 15

14

addition of the EDC and NHS the solution became turbid but no precipitation was evident. Both 17

16

the EDC and NHS were dissolved in ethanol to limit instability due to hydrolysis while 18 19

maintaining polymer solubility. 20 21 2 23

O

H3C Cl H N+ CH3

24 25

H3C + H N CH3

OH

26 27

H3C O

HN

NH2

HN

HN

HN

NH2

NH2

+

O

N

28

NH

H3C O

O H2N

O NH

NH

CH3

O

NH O

NH

OH

NH O

N

29

CH3

32

31

Scheme 1. 3

HN

HN

O NH

NH

O

O

O

NH

NH O

OH

NH O

HN

HN

30

H3C

CH3

NH2

HN

O

O NH

HN

HN

HN

N

+

NH2

HN

NH2

NH2

Peptide crosslinking reaction scheme. Carboxylic acid groups on the

35

34

poly(methacrylic acid-co-N-vinylpyrrolidone) linear polymer are activated by EDC, then react 37

36

with at least two of the five primary amine groups on the GRRRGK peptide to form a 38 39

crosslinked hydrogel network. 40 41 43

42

After activation of the carboxylic acid groups, the pH was raised to ~7-8 to facilitate attack on 45

4

the primary amines of the oligopeptide 47

46

44

. Various polymer:EDC:NHS:peptide weight ratios

were tested to maximize peptide incorporation. All formulations with peptide content below a 48 50

49

polymer:peptide ratio of ~3:1 failed to produce hydrogels. A best performing formulation of 52

51

solvents, polymer, and EDC-NHS based on reaction efficiency and reproducibility was an 53 54

ethanol-water mixture with a polymer:EDC:NHS:peptide weight ratio of 20:10:3.3:10. 5 56 57 58 59 60 ACS Paragon Plus Environment

12

Page 13 of 41

Biomacromolecules

1 3

2

The peptide was added at a molar ratio of 1:3.6 relative to the EDC; the free amine groups 5

4

were in 1.4x excess relative to the theoretical maximum of activated carboxyl groups. At this 6 8

7

feed ratio, the degree of crosslinking is expected to be about 3. Upon addition of the peptide 10

9

solution the mixture was immediately turbid and precipitation of crosslinked polymer was 1 12

evident. After reacting for at least 8 hours the crosslinked polymer typically resembled an 13 15

14

amorphous hydrogel. 17

16

Following washes and lyophilization the hydrogel appeared as fluffy white chunks, as seen in 18 19

Figure S.1 (initial). The dried hydrogel was easily crushed into a powder consisting of particles 20 2

21