Supramolecular Gels Derived from Simple Organic Salts of Flufenamic

5 days ago - Supramolecular Gels Derived from Simple Organic Salts of Flufenamic acid: Design, Synthesis, Structures and Plausible Biomedical ...
0 downloads 0 Views 883KB Size
Subscriber access provided by UNIV AUTONOMA DE COAHUILA UADEC

Characterization, Synthesis, and Modifications

Supramolecular Gels Derived from Simple Organic Salts of Flufenamic acid: Design, Synthesis, Structures and Plausible Biomedical Application Rumana Parveen, Bandi Jayamma, and Parthasarathi Dastidar ACS Biomater. Sci. Eng., Just Accepted Manuscript • DOI: 10.1021/acsbiomaterials.9b00153 • Publication Date (Web): 01 Apr 2019 Downloaded from http://pubs.acs.org on April 5, 2019

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 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 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.

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 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Supramolecular Gels Derived from Simple Organic Salts of Flufenamic acid: Design, Synthesis, Structures and Plausible Biomedical Application.

Rumana Parveen, Bandi Jayamma and Parthasarathi Dastidar* AUTHOR ADDRESS: School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S .C. Mullick Road, Kolkata700032,West Bengal India, E-mail: [email protected]; Fax: 2473-2805; Tel: +91-33-2473-4971.

ACS Paragon Plus Environment

+91-33-

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 38

ABSTRACT. Following supramolecular synthon rationale in the context of crystal

engineering,

a

non-steroidal-anti-inflammatory-drug

(NSAID)

namely Flufenamic acid (FA) and its -alanine monopeptide derivative (FM) were converted to a series of primary ammonium monocarboxylate (PAM) salts. Majority of the PAM salts (~90 %) showed gelation with various

solvents

including

water

and

methyl

salicylate

(important

solvents in topical gel formulation). Structure-property correlation studies based on single crystal X-ray diffraction (SXRD) and powder Xray diffraction (PXRD) data provided intriguing insights into the structure of the gel network. Furthermore, one of the gelator salts (S7) displayed anti-cancer activity on a highly aggressive human breast cancer

cell

line

(MDA-MB-231)

as

revealed

by

MTT,

PEG2

and

cell

migration assays.

KEYWORDS: Anti-inflammatory; Anti-cancer; Cell-migration; SAFiNs; Supramolecular gels.

INTRODUCTION.

Supramolecular

gels1-8

are

fascinating

class

of

viscoelastic (solid-like soft) materials usually formed by dissolving a little amount of solid (gelator) in a suitable solvent (gelling solvent) followed by external stimuli such as heat, photo-irradiation, sound, change in pH etc. Supramolecular

gelators, especially low

molecular weight gelators (LMWGs, a class of small molecules having molecular weight < 3000) are increasingly becoming important in material science due to their intriguing potential applications in developing electro-optics/photonics,9,10

sensors,11,12

cosmetics,13

structure

directing agents,14 reaction media for catalysis,15 conservation of ACS Paragon Plus Environment

Page 3 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

art,16 bio-materials17-24 etc.

However, wide structural diversity of

gelator molecules and lack of clear understanding of gelation mechanism in molecular level (despite systematic efforts made by various groups2530)

make it difficult to design a supramolecular gelator a priori. One

of the early attempts to design a gelator molecule was based on structure-property correlation approach proposed by Shinkai et al.;31 based on a few single crystal structures of sugar derivatives and their property (gelation/non-gelation), they proposed that molecules prone to form 1D hydrogen bonding networks (HBNs) were expected to promote gelation as such 1D self-assembly of gelator molecules would promote the formation of network of fibers (known as self-assembled fibrillar networks or SAFiNs)32 that eventually immobilized the solvent molecules due to surface tension or capillary force action resulting in gel. Our group

has

been

supramolecular

developing

synthon33

supramolecular

approach

in

gelators

the

context

following of

crystal

engineering.34 Such link between crystal engineering and gels was first introduced by us and provided a way to occasional prediction of gelation as well as clear understanding of structure landscape of such selfassembled materials.35 Based on more than hundreds of crystal structures of

organic

supramolecular

salts,36 synthons

we

identified

namely

four

Primary

major

Ammonium

gel-inducing

Monocarboxylate

(PAM),37 Secondary Ammonium Monocarboxylate (SAM),38 Secondary Ammonium Dicarboxylate

(SAD),39 and

Primary

Ammonium

Dicarboxylate

(PAD).40

Amongst these, PAM synthon is found to be the most promising gelinducing

supramolecular

synthon.

There

are

several

advantages

associated with organic salts for developing LMWGs; a) salt formation ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 38

is the easiest reaction to carry out, b) purification step does not include time consuming and expensive column chromatography, c) yield is often near quantitative, d) 100 % atom economy makes the synthesis of the salts green and sustainable, e) virtually infinite combination of commercially available reactants (acids and amines) allow one to generate a large library of organic salts for gelation sacnning. In the present work, we exploited PAM synthon to develop organic salt based

gelators

for

self-drug-delivery

applications.

Self-drug-

delivery41 is an alternative approach to conventional drug-delivery wherein

a

drug

delivery

vehicle

(such

as

vesicles,42 polymer

gel

matrix,43 reverse micelles44 etc.) is not needed to deliver the drug; instead, it can be delivered to the target site by itself (hence the name self-drug-delivery). In this approach, one can avoid the issues related to delivery vehicles (syntheis, cytotoxicity, biodegradability and biostability of the vehicle, drug loading and release kinetics etc.). Self-drug-delivery45 can be achieved by modifying a drug to a supramolecular gelator (without compromising its drug action) so that a suitable gel derived from the gelator drug can be applied either via non-invasive (topical) or invasive (subcutaneous) route.

ACS Paragon Plus Environment

Page 5 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Scheme 1. An overview of the results described in the present work. For example, a long acting implant in the form of a supramolecular hydrogel

derived

from

a

synthetic

octapeptide

hydrogelator

was

installed via subcutaneous route to treat a growth disorder namely acromagley.46 Pyrene derivative of a highly potent antibiotic namely vencomycin

was

reported

to

form

hydrogel

that

displayed

enhanced

antibiotic activity.47 D-glucosamine derivative hydrogel was effective in treating wound in in vivo studies.48 We have also demonstrated that supramolecular synthon approach was helpful in coverting non-steroidalanti-inflammatory drugs (NSAIDs) to supramolecular gels that could be used to treat inflammation (both in vitro49 and in vivo50). Flufenamic acid (FA) is a well known NSAID containing a reactive monocarboxylic acid functionality. The drug is practically insoluble in water causing poor bioavailability and if administered orally, it causes severe gastrointestinal side effects. Having this bacground in mind, we synthesized a series of PAM salts of FA and its -alanine derivative (FM) by reacting with a number of primary amines with the hope of obtaining gelator salts that could be useful in self-drugACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

delivery

applications

via

topical

route.

Page 6 of 38

Synthesis

of

the

salts,

gelation studies, structure-property (gelation) correlation based on single crystal and powder X-ray diffraction data of some of the salts, biocompatibility (MTT assay), anti-inflammatory property (PGE2 assay) and anti-cancer property (cell migration assay) of some of the selected salts are discussed in the context of developing self-drug-delivery system (Scheme 1).

Experimental Section Materials and Methods: All the chemicals including Flufenamic acid and various amines were purchased from Sigma Aldrich and used without further purification. Solvents were AR (Analytical Reagents) grade and were used without any further purification.

Mouse macrophage RAW 264.7 and human breast

cancer cells MDA-MB-231 were purchased from the American Type Culture Collection (ATCC). For PGE2 assay Prostaglandin E2 EIA Kit – Monoclonal (Cayman Chemicals, Ann Arbor, MI) was used. Methods: NMR spectra (both 1H and

13C)

were recorded using both 400 MHz and 500

MHz spectrometer (Bruker Ultrashield Plus-500). FT-IR spectra were recorded

using

Microscopy

(SEM)

instrument was

FTIR-8300,

performed

in

a

Shimadzu. JEOL,

Scanning

JMS-6700F.

Electron

Rheological

experiments were carried out using MCR 102 Anton Paar modular compact rheometer. MTT and PGE2 assay were performed using a multi plate ELISA reader (Varioskan Flash Elisa Reader, Thermo Fisher. For

capturing

images of the cells, Carl Zeiss axio observer z1 fitted with a Hamamatsu ACS Paragon Plus Environment

Page 7 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

orca flash 4.0 was used. Fluorescence measurements were done with a Horiba Jobin Yvon Fluoromax-4 spectrofluorometer.

Synthesis of -alanine monopeptide of flufenamic acid (FM) First benzotriazole (357mg, 3mmol) was taken in

dry DCM (20ml) at

room temperature. To this solution thionyl chloride (90µl, 1.25 mmol) was added gradually and the reaction mixture was stirred for 20 minutes. The mother drug flufenamic acid (281 mg, 1mmol) was dissolved in dry THF (15 ml) and added dropwise to the first reaction mixture. The mixture was then stirred for 15 hours at room temperature. The white precipitate, thus obtained, was filtered off. Filtrate was concentrated under reduced pressure to obtain benzotriazole derivative of flufenamic acid as a white solid mass which was then dissolved in THF (20 ml). βalanine (89 mg, 1mmol ) and triethylamine (100 µl) dissolved in H2O and THF (20 ml, 1:4 ratio, v/v) was poured into the solution of bezotriazole derivative

of

flufenamic

acid

and

stirred

for

24

hours

at

room

temperature followed by removal of THF under reduced pressure. The concentrated solution was acidified by HCl (1N) in order to obtain a white precipitate. The white ppt was filtered and washed 3-4 times with distilled water in order to remove the excess acid. The solid product was then dried in a vacuum desiccator and was further purified by column chromatography (using 3% MeOH in DCM) to obtain ~61%) (Scheme 2).

ACS Paragon Plus Environment

pure product FM (yield

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 38

Scheme 2. Synthetic procedure of -alanine monopeptide of flufenamic acid (FM). Synthesis of salts All the salts were prepared by reacting the corresponding acids and amines in 1:1 molar ratio in methanol at room temperature. Solid mass was obtained after the evaporation of the solvent in near-quantitative yield. FM:

1H

NMR (400 MHz, DMSO-d6, 25°C): δ=12.22(s,1H), 9.59(s, 1H),

8.59(s, 1H), 7.63-7.61(d, J= 8 Hz, 1H),7.48-7.44(t, J= 8Hz, 1H), 7.387.36(d, J= 8 Hz 1H),7.33-7.31(d, J= 8Hz, 3H), 7.21-7.19(d, J= 8 Hz, 1H), 6.96-6.95(t, J=8 Hz, 1H), 3.44-3.31(m, 2H), 2.49-2.45(t, J= 8Hz, 2H).

13C

NMR (100MHz, DMSO-d6 25°C):172.79, 168.31, 143.10, 142.30,

131.75, 130.37, 128.95, 121.54, 121.31, 119.84, 117.01, 116.97, 116.77, 113.93, 35.33, 33.50 ppm (Figure S1 in supporting information). FT-IR (KBr pellet): 1709 cm-1 (>C=O stretching for –COOH, Figure S12 in supporting information). S1:

1H

NMR (400 MHz, DMSO-d6, 25°C): δ=7.94-7.92(d, J= 8 MHz, 1H),

7.46-7.42(t, J= 8MHz, 2H), 7.39-7.21(m, 8H), 6.78-6.74(t, J=8Hz, 1H), 3.06-3.02(t, J= 8Hz, 2H), 2.90-2.85(t, 2H, J=8Hz).

13C

NMR (100MHz,

DMSO-d6, 25°C): 171.11, 143.96, 143.81, 137.66, 132.11, 130.51, 130.42, ACS Paragon Plus Environment

Page 9 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

130.03, 128.68, 128.62, 126.70, 125.65, 123.71, 121.25, 118.40, 116.21, 114.28, 113.35, 40.18, 33.46 ppm (Figure S2 in supporting information). FT-IR (KBr pellet): 1605cm-1 (>C=O stretching for COO-, Figure S12 in supporting information). S2: 1H NMR (400 MHz, DMSO-d6, 25°C): δ= 8.16-8.14(d, J=8Hz, 1H), 7.997.92(m, 3H), 7.63-7.56(m, 3H), 7.54-7.50(t, J=8Hz, 1H), 7.47-7.44(t, J=8Hz, 1H), 7.40-7.38(d, 2H),

7.17-7.15(d,

J=8Hz, 1H), 7.34(s, 1H), 7.26-7.24(d, J=8Hz,

J=8Hz, 1H), 6.79-6.75(m, 1H), 4.51(s, 2H).

13C

NMR

(100MHz, DMSO-d6, 25°C): 170.74, 144.13, 143.60, 133.28, 132.07, 130.76, 130.49, 130.31, 130.00, 128.83, 128.66, 126.89, 126.68, 126.20, 125.42, 123.48, 122.91, 121.49 118.40, 116.38, 113.41, 113.63, 40.43, 38.88 ppm FT-IR (KBr pellet): 1616 cm-1

(Figure S3 in supporting information).

(>C=O stretching for COO-, Figure S12 in supporting information). S3:

1H

NMR (400 MHz, DMSO, 25°C): δ= 7.97-7.94(d, J=7Hz, 1H), 7.56-

7.54(d, J=8Hz, 1H), 7.38-7.35(d, J=8Hz, 4H), 7.30-7.26(t, J=7.2 Hz, 2H), 7.14-7.01(m, Hz,

4H), 6.83-6.79(t,

2H), 3.13-3.07(t, J=7.2 Hz,

J=8Hz, 1H), 3.23-3.20(t, J=6.8

2H),.

13C

NMR (100 MHz, MeOD, 25°C):

175.62, 145.60, 138.34, 133.20, 132.71, 132.39, 132.18, 131.05, 128.17, 124.25, 123.86, 123.06, 122.76, 120.05, 119.79, 118.87, 118.05, 115.81, 112.55, 110.33, 41.27, 40.42, 24.61 ppm (Figure S4 in supporting information). FT-IR (KBr pellet): 1620 cm-1 (>C=O stretching for COO-, Figure S12 in supporting information). S4:

1H

NMR (400 MHz, DMSO-d6, 25°C): δ=7.95-7.93(d, J=8Hz, 1H), 7.49-

7.45(t, J=8Hz, 1H), 7.42-7.40(d, J=8Hz, 1H), 7.35(s, 2H), 7.26(s, 1H), 7.17-7.15(d, J=8Hz, 1H), 6.79-6.75(t, J=8Hz, 1H), 3.61-3.58(m, 4H), 3.09-3.07(t, J=4Hz, 1H).

13C

NMR (100MHz, DMSO-d6, 25°C): 171.13, 143.99,

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 38

143.75, 132.11, 130.45, 129.98, 125.59, 122.89, 121.41, 118.32, 116.28, 114.19, 113.52, 59.12, 59.03, 54.30 ppm (Figure S5 in supporting information). FT-IR (KBr pellet): 1625 cm-1 (>C=O stretching for COO-, Figure S12 in supporting information). S5: 1H NMR (500 MHz, DMSO-d6,

25°C): δ= 7.92-7.91(d, J=7.5Hz, 1H),

7.45-7.42(t, J=8.5Hz, 1H), 7.38-7.36(d, J=7.5Hz, 1H), 7.31(s, 1H), 7.24-7.19(m, 2H), 7.13-7.11(d, J=10Hz, 2H), 6.75-6.72(t, J=7.5Hz, 2H),

1H), 6.86-6.83(t, J=7.5Hz,

J=7 Hz, 2H), 3.71-3.69(d, J=7.5Hz, 6H), 3.01-2.97(t,

2.78-2.75(t,

J=7.5Hz, 2H).

13C

NMR (100MHz, DMSO-d6,

25°C): 170.92, 148.76, 147.57, 143.89, 131.98, 130.37, 130.06, 125.29, 123.84, 123.13, 121.06, 120.53, 118.19, 115.95, 114.11, 113.15, 112.56, 112.01, 55.50, 40.31, 33.24 ppm (Figure S6 in supporting information). FT-IR (KBr pellet): 1600 cm-1 (>C=O stretching for COO-, Figure S12 in supporting information). S6:

1H

NMR (400 MHz, DMSO-d6, 25°C): 9.69(s, 1H), 8.65(S, 1H), 7.61-

7.59(d, J=8Hz,1H), 7.46-7.42(t, J=8Hz, 1H), 7.37-7.25(m, 6H), 7.217.17(t, J=8Hz, 4H), 6.92-6.89(t, J=8Hz, 1H),

3.38-3.37(d, J=5.2Hz,

2H), 2.92-2.89(t, J=8Hz, 2H), 2.80-2.76(t, J=8Hz, 2H), J=7Hz,

2H).

13C

2.28-2.24(t,

NMR (100MHz, DMSO-d6 25°C): 174.84, 168.13, 143.25,

142.39, 138.56, 131.72, 130.49, 128.86, 128.68, 128.52, 126.42, 125.57, 122.86, 121.68, 36.58, (KBr

36.02,

pellet):

120.00, 117.06, 116.80,

113.99,

41.08,

40.13,

35.30 ppm (Figure S7 in supporting information). FT-IR 1636

cm-1

(>C=O

stretching

for

COO-,

Figure

S12

in

supporting information). S7:

1H

NMR (400 MHz, DMSO-d6, 25°C): 9.63(s, 1H), 8.64(s, 1H), 8.12-

8.10(d, J=8Hz, 1H), 7.95-7.93(d, J=8Hz, 1H), 7.84-7.82(d, J=8Hz, 2H), ACS Paragon Plus Environment

Page 11 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

7.63-7.62(d, J=8Hz, 2H), 7.56-7.54(d, J=8Hz, 3H), 7.53-7.50(d, J=8Hz, 1H), 7.48-7.46(t, J=8Hz 1H), 7.40-7.32(m, 1H), 7.22-7.20(d, J=8Hz, 2H), 6.97-6.93(t, J=8Hz, 3H), 4.28(s, 2H), 2.42-2.38 (t, J=8Hz, 2H).

13C

NMR

(400MHz, DMSO-d6, 25°C): 170.93, 168.30, 148.76, 147.57, 143.89, 131.98, 130.38, 130.06, 125.29, 123.84, 123.13, 121.06, 120.53, 118.19, 115.95, 114.11, 113.15, 112.56, 112.00, 55.50, 40.31, 33.24ppm (Figure S8 in supporting information). FT-IR (KBr pellet): 1639 cm-1 (>C=O stretching for COO-, Figure S12 in supporting information). S8:

1H

NMR (400 MHz,

7.59(d, J=8Hz,

DMSO-d6, 25°C): 9.68(s, 1H), 8.72(s,1H), 7.61-

1H), 7.53-7.51 (d, J=8Hz,

1H), 7.47-7.43(t, J=8Hz,

1H), 7.39-7.30(m, 5H), 7.20-7.16(t, J=8Hz, 2H), 7.08-7.04(m, 1H), 6.966.89(m, 2H), 3.38-3.35(t, J=8Hz, J=8Hz, 2H). 142.34,

13C

2H), 2.94-2.89(m,

4H), 2.30-2.26(t,

NMR (400MHz, DMSO-d6, 25°C): 174.16, 168.09, 143.21,

136.33,

131.70, 130.46, 130.32, 130.00, 128.84, 127.05,

123.08, 121.61, 121.06, 119.97, 118.37, 118.21, 116.99, 116.78, 114.01, 111.49, 110.74, 40.52, 36.35, 35.54, 25.51 ppm (Figure S9 in supporting information).

FT-IR (KBr pellet): 1624 cm-1 (>C=O stretching for COO-,

Figure S12 in supporting information). S9: 1H NMR (400 MHz, DMSO-d6, 25°C): δ=9.65(s, 1H), 8.62(s, 1H), 7.617.59(d, J=8Hz, 1H), 7.47-7.44(t, J=8Hz, 1H), 7.38-7.29(m, 4H), 7.207.18(d, J=8Hz, 1H),

6.94-6.90(t, J=8Hz, 1H),

3.38(t, J=4Hz, 4H), 2.91(s, 1H), 2.28(s, 2H).

13C

3.49(s, 2H),

3.41-

NMR (400MHz, DMSO-d6,

25°C): 174.68, 168.19, 143.25, 142.38, 131.77, 130.51, 130.06, 128.92, 125.57, 121.65, 120.04, 117.08, 116.86, 114.03, 69.85, 60.55, 54.43, 36.35, 35.59 ppm (Figure S10 in supporting information).

ACS Paragon Plus Environment

FT-IR (KBr

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 38

pellet): 1637 cm-1(>C=O stretching for COO-, Figure S12 in supporting information). S10: 1H NMR (500 MHz,

DMSO-d6, 25°C): 9.68(s,

1H), 8.69(s, 1H), 7.60-

7.56(d, J=7.5Hz, 1H), 7.46-7.43(t, J=7.5Hz, 1H), 7.38-7.30(m, 3H), 7.19-7.18(d, J=7.5Hz, 1H), 6.98-6.89(m, 3H), 6.67-6.66(d, J=8.5Hz, 2H), 3.37-3.36(d, J=5.5Hz, 2H), 2.79(s, 2H), 2.63-2.59(t, J=7.5Hz, 2H), 2.29-2.26(t, J=7Hz, 2H).

13C

NMR (400MHz, MeOD, 25°C): 171.29, 163.65,

150.90, 149.99, 144.88, 144.11, 132.99, 131.18, 130.55, 129.96, 124.27, 123.29, 122.88, 122.22, 121.39, 118.54, 118.28,

115.59,

113.73,

113.53, 56.56, 42.07, 37.48, 35.52, 34.22 ppm (Figure S11 in supporting information).

FT-IR (KBr pellet): 1623 cm-1 (>C=O stretching for COO-,

Figure S12 in supporting information).

Measurement of Tgel Gel-sol dissociation temperature (Tgel) was measured by dropping ball method; a glass ball (400 mg) was placed on a 0.5 ml gel taken in a test tube (internal diameter = 1.0 cm) which was then immersed in an oil bath and the temperature was gradually increased till the ball touched the bottom of the test tube. The temperature at which the ball touched the bottom of the test tube was recorded as Tgel. Microscopy The SEM images were prepared by smearing a small amount of gel on SEM stub followed by drying overnight at room temperature. Single crystal X-ray diffraction SXRD data were obtained using MoKα (λ = 0.7107 Å) radiation on a BRUKER APEX II diffractometer equipped with CCD area detector. Data ACS Paragon Plus Environment

Page 13 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

collection, data reduction, structure refinement were performed using the software package of SMART APEX-II. All structures were solved by direct method and routinely refined. Non-hydrogen atoms were treated anisotropically except for the disordered atoms. Flourine atoms of – CF3 in S2, S5 and S8 were found to be disordered over two positions. Wherever possible, the hydrogen atoms were located on difference Fourier map and refined; in other cases, the hydrogen atoms were geometrically fixed. MTT Assay Cytotoxicity of the gelator salts and the mother drug flufenamic acid was determined in RAW 264.7 as well as MDA-MB-231 cells using MTT (3(4,

5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium

Cells

were

cultured

in

Dulbecco’s

Modified

bromide)

Eagle’s

assay.

Medium

(DMEM)

supplemented with 10% Fetal Bovine Serum (FBS), 1% penicillin and streptomycin in a humidified incubator at 37° C and 5% CO2. In a typical experminent,

the

cells

were

grown

in

96-well

plates

(0.5

×

104

cells/well). After 24 hours, the cells were charged with various concentrations (upto 1 mM) of the gelators or DMEM alone (control) for 72 h in humidified incubator at 37˚C and 5% CO2. Then, the culture medium was replaced with 100 μg of MTT per well and kept the solution for 4 h at 37˚C. Thus formazan was produced by the mitochondrial reductase from live cells which was further dissolved by DMSO (100 μL/ well). The cells were incubated for 40 minutes at room temperature. The colour

intensity

of

the

formazan

solution

was

measured

using

a

multiplate ELISA reader at 570 nm (Varioskan Flash Elisa Reader, Thermo Fisher)., which was directly correlated to the cell viability or amount ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 38

of living cell, The percentage of live cells in gelator-treated sample was thus calculated considering DMEM-treated cells as 100%. PGE2 assay PGE2 assay of S7 was carried out on both Raw 264.7 cells and human breast cancer cells MDA-MB-231. Approximately 1× 106 cells/well was seeded in a six well plate and kept it in a humidified incubator at 37°C saturated with CO2for over night. DMEM (1 ml) was taken in one well which was used as control. For Raw 264.7 to obtain maximum inflammation, the cells (rest wells) were charged with inflammatory agents such as lipopolysaccharid (LPS) and interferon-gamma (IFN-γ) so that final concentrations were 1μg/mL and 100 ng/mL, respectively. Out of

these

inflammated

cells,

two

wells

were

treated

with

0.1

mM

flulfenamic acid and S7 respectively in such a way that the total culture medium (DMEM) volume in each of the cases remained 1 ml. Next, it was kept for 24 hrs at 37°C saturated with CO2. Cell soup was then added to PGE2 kit. After 24 hours, the amount of PGE2 produced was determined using ELMAN’s reagent. For human breast cancer cells MDAMB-231, as the cells were already inflammated, no inflammatory agent such as LPS and IFN-γ was added. Cell imaging For cell imaging studies, MDA-MB-231 cells were cultured in 9 cm2 culture plate and kept it in a humidified incubator at 37°C saturated with CO2 for 24 hours. S7 (0.1 mM) was then added to it and incubated for 15 minutes. The cell medium was sucked out and the MDA-MB-231 cells were treated with 4% paraformaldehyde for 2-3 minutes for cell fixation.

ACS Paragon Plus Environment

Page 15 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

The cancer cells were then washed with 1X PBS. The fluorescence images were captured using 20X objective lens using fluorescence microscope.

Results and Discussion Salts depicted in Scheme 3 were synthesized by reacting FA and FM with the corresponding amines in 1:1 molar ratio in MeOH at room temperature. The resulting white solids after evaporation of the solvent were subjected to FT-IR. Disappearance of –COOH stretching band at 1660 and 1709 cm-1 and appearance of –COO- stretching band at ~1600-1625 and ~1624-1639 cm-1 for FA and FM, respectively confirmed salt formation. The purity of the salts was also supported by 1H and

13C

NMR (Experimental

section). Remarkably most of the salts (9 out of 10 salts) were found to arrest the flow of various selected solvents indicating gel formation (Table 1). The gels were thermo-reversible over a few cycles. While the minimum gelator

concentration

(MGC)

varied

from

~2.1-4

wt

%,

the

gel

dissociation temperature (Tgel) was found to be moderate (49-73 C). Amongst the gelator salts, S4 and S5 were found to be ambidextrous as it produced both hydro- and organo-gels. While S1, S4 and S5 showed gelation with pure water (hydrogel), S5 and S7 were found to form gel with methyl salicylate (MS) as well. Since MS is an important ingredient of several commercially available anti-inflammatory topical gels, the MS gels derived from S5 and S7 are particularly important in the present context.

As the main purpose of the present study is to develop ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 38

supramolecular topical gel for developing self-drug-delivery system, the hydrogels and MS gels derived from S1, S4, S5 and S7 were studied further.

Scheme 3. Various PAM salts of FA and FM studied herein. The fact that various non-covalent interactions were crucial for gelation was clearly established from the steady increase of Tgel followed by a plateau with the increase in gelator concentration (Figure 1). Such table top rheological response was typical for supramolecular gels.51 Morphological

features

of

the

gel

network

were

revealed

in

SEM

micrograph. In a typical experiment, SEM samples were prepared by smearing a little amount of gel (at MGC concentrations) on a SEM stub followed by drying under room temperature for 12 hours. In all the cases, highly entangled 1D fiber like morphology was observed (Figure 2). ACS Paragon Plus Environment

Page 17 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Figure 1: Tgel vs [gelator] (wt % w/v) plot for hydrogel of S1, S4 and S5

and the MS gels of S5 and S7.

Flow properties of the gels (4.0 wt % hydrogels of S1, S4 and S5 and MS gels of S5 and S7) were studied by dynamic rheology. Typical viscoelastic response (wherein elastic modulus (Gˊ) is significantly larger than the viscous modulus (Gˊˊ) and remain frequency invariant over a longer time scale in frequency sweep experiment) is observed for all the gels under study confirming true viscoelastic nature of the gels (Figure 3).

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 38

Figure 2: Physical appearance of the gels (inset) and morphological feature of the gel network in SEM images; hydrogels of a) S1, b) S4 and c) S5 and MS gels of d) S5 and e) S7.

Figure 3: Rheological responses (frequency sweep) of the 4.0 wt % (w/v) hydrogels of a) S1, b) S4 and c) S5 and MS gels of d) S5 and e) S7 at a given constant strain of 0.1 % at 25°C.

Rheoreversibility indispensable

for

is

an

important

developing

gels

for

material

property

self-healing.52

that and

is

drug-

delivery53 applications. If a gel is rheoreversible, it should display gel-sol-gel transitions over a few high-low strain cycles. In other words, the gel under study should display viscus response (Gˊˊ>Gˊ) under high strain condition and quick elastic response (Gˊˊ>Gˊ) when the strain is removed (relaxation). This property is particularly important if the gel is to be administered topically or subcutaneously. Two selected gels (4 wt % hydrogel of S1 and MS gel of S7) displayed ACS Paragon Plus Environment

Page 19 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

excellent

rheoreversible

behaviour

when

they

were

subjected

to

a

constant strain of 20 % (much higher than the corresponding critical strain - 7.5 and 4.4 % for hydrogel of S1 and MS gel of S7, respectively; Figure S13, supporting information) and allowed to relax for 100s (Figure 4).

Figure 4: Oscillatory shear of 4 wt%

a) hydrogel of S1 and b) MS gel

of S7. Structure-property correlation: To get an insight into the role of supramolecular synthons present in these salts and their influence on gelation, we set out to determine the single crystal structures of the salts. Single crystals were obtained for the gelator salts S1, S2, S4, S5 and S8 and subjected SXRD data collection (Table s1). Analysis of the crystal structures revealed that S1, S2, S5 and S8 displayed 1D HBN. On the other hand, S4 showed 2D HBN mainly because of the participation of the hydrogen bond functionalized cationic moiety in the salt. Closer look at the crystal structures showed that S1 possessed a 1D cylindrical HBN instead of a typical PAM synthon (‘X’ or ‘W’); PAM synthon ‘X’ was present in S2, whereas S8 displayed PAM synthon ‘W’ despite having an additional hydrogen bonding functionality (indole NH) that participated in hydrogen bond with the O atom of the carbonyl ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 38

group of adjacent peptide salt; an overall 1D HBN network was observed in S5 despite having lattice occluded water molecules that participated in hydrogen bonding interactions with the carboxylate O and ammonium N atoms; the only gelator salt that displayed 2D HBN was S4; absence of a typical PAM synthon (‘X’ or ‘W’) in this structure could be due to the additional hydrogen bonding moieties (two hydroxyl group of the cationic species) that participated in hydrogen bonding (Figure 5). Powder X-ray diffraction (PXRD) patterns (simulated, bulk solid and xerogel) showed nice correspondence with respect to the peak positions thereby indicating high crystalline phase purity of the bulk as well as revealed the structure of the gel networks in their xerogel state (Figure 6). Thus, SXRD and PXRD data clearly supported Shinkai’s hypothesis

and

validate

our

supramolecular

synthon

approach

for

designing supramolecular gelators derived the NSAID Flufenamic acid.

ACS Paragon Plus Environment

Page 21 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Figure 5: Crystal structure illustration of the salts; a) cylindrical 1D HBN in S1, b) synthon ‘X’ in S2, c) synthon ‘W’ in S8, d) lattice occluded water mediated 1D HBN in S5 and e) 2D HBN S4 (moieties not participating in HBN are hidden for clarity).

Figure 6: X-ray powder diffraction patterns under various conditions. Biological studies: Owing to the gelation ability of S1, S4, S5 and S7 with solvents like water

and

methyl

salicylate

(both

suitable

for

biomedical

applications), we first evaluated the cytotoxicity profile of these salts in a macrophage cell line namely RAW 264.7 by MTT assay54 for 72 hours at 37 °C in Dulbecco's Modified Eagle's Medium(DMEM). The IC50 values for S1, S4, S5, S7 and FA were 0.1, 0.1, 0.25, 0.25 and 0.1 mM, respectively.

Cell viability of S7 was maximum ( % of survival of S7

at 0.25 mM concentration was ~65% whereas for S5 that was ~56%)

and

therefore, was best suited for further biological studies (Figure 7).

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 38

Figure 7: MTT assay of a) S1, b) S4, c) S5, d) S7 and e) FA in RAW 264.7 cell line for 72 hrs. at 37˚C.

It may be mentioned that NSAIDs reduce inflammation by inhibiting PGE2 producing enzymes like cyclooxygenase (COX-1/COX-2). PGE2 assay allows one to measure quantitatively extra-cellular concentration of PGE2; the less the concentration of PGE2, the better is the anti-inflammatory response. We, therefore, carried out prostaglandin E2 (PGE2) assay of the most biocompatible salt S7 in RAW 264.7 cell line following a published protocol55 in order to assess its anti-inflammatory response compared to that of the parent drug FA (see experimental section for details).

External

inflammation-inducing

agents

such

as

lipopolysaccharide (LPS) and interferon-gamma (IFN-γ) were gradually added to obtain maximum inflammation in the cells as measured by the concentration

of

PGE2

in

the

cell

media.

After

24

hours,

PGE2

concentration (2006 pg/mL) was found to be significantly higher in the inflammated cells than that (614 pg/ml) in the control cells (without LPS and IFN-γ) under identical conditions. PGE2 concentration in cell media was significantly reduced to 45 pg/mL and 88pg/mL by S7 (0.1 mM) and FA (0.1 mM), respectively indicating the gelator salt S7 did possess ACS Paragon Plus Environment

Page 23 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

anti-inflammatory property which was even better than that of the mother drug FA (Figure 8).

Figure 8: Anti-inflammatory response of FA and S7 in RAW 264.7 cells.

Anti-cancer activities: It has been reported that chronic inflammation at a particular site may cause cancer.56,57 Increased level of concentration of PGE2 has been detected in breast cancer patients58-60 PGE2 is also shown to play important role in progression and metastasis of cancer cells61 Tumor growth and metastasis are also linked to PGE262

Growth and progression

of prostate cancer has been successfully retarded by a combination therapy involving a well-known NSAID namely naproxen and vitamin D3.63 Recent in vitro studies revealed that a naproxen derivative was able to inhibit the cell migration of human breast cancer cells.64 Keeping these backgrounds in mind, we then evaluated the anti-cancer behaviour of the gelator salts S1, S4, S5, S7 and compared with that of the parent drug FA by MTT assay in a highly aggressive human breast cancer cell line MDA-MB-231; the results suggested that S7 had the best ability to ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 38

kill the cancer cells (IC50 0.1 mM) which was even better than that of the mother drug (IC50 0.25 mM) (Figure 9).

Figure 9: Cell viability assay (MTT assay) in human breast cancer cells MDA-MB-231 at 37° C for 72 h in presence a) S1, b) S4, c) S5, d) S7 and e) FA, respectively.

Anti-cancer property of S7 was further supported by its ability to reduce extra-cellular concentration of PGE2 in the same cell line as revealed by PGE2 assay; after 24 hours, the amount of PGE2 production was 466 pg/ml for non-treated cells which was significantly reduced when the cells were charged with FA (24 pg/ml) and S7 (16 pg/ml) in separate experiments (Figure 10).

Figure 10: PGE2 assay of FA and S7 in MDA-MB-231.

ACS Paragon Plus Environment

Page 25 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Cell migration assay: Uncontrolled migration of cells causing metastasis in cancer patients is a common phenomenon. Thus, in vitro inhibition of cancer cell migration (cell migration assay) is a measure of anti-cancer behaviour of any substance. We have thus subjected the gelator salt S7 to cell migration assay64 on the same breast cancer cell line i.e. MDA-MB-231. In a typical experiment, cells were seeded in a 6 well plate in DMEM. After 24 hours, a scratch was introduced on it by a sterile pipette tip (200µl). The cells were then charged with 0.1 mM of FA and S7. Control experiment was also performed where no drug was added. Times resolved still images were captured for 24 hours to measure the migration speed of the cells. After 24 hours, cells covered 40% of the gap for FA, 23% for S7 whereas in the control experiment, 84% gap was covered. The cell migration speed was 8.3, 3.7 and 2.2 µm/h in control, FA, S7 treated cells, respectively, suggesting that S7 was able to inhibit effectively the migration of the cancer cells (Figure 11).

ACS Paragon Plus Environment

.

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 38

Figure11: Migration of the cell front observed at different time intervals in scratch assay performed on MDA-MB-231 cells. Cellular imaging: Finally, cell imaging experiment was performed in order to probe whether

the

modified

gelator

salt

was

internalized65

or

not.

Interestingly, S7 was internalized in MDA-MD-231 cells as revealed by the green luminescence when it was excited with blue light (ex = 488 nm) under a fluorescence microscope (Figure 12). Green fluorescence (em = ~539 nm) was also observed from the DMSO solution of S7 (ex = 488 nm)(Figure S24).

Figure 12: (a) Bright field image; (b) Fluorescence image; (c) overlay image of S7 (using 0.25 mM concentration)

in MDA-MB-231 cells.

Conclusions Thus, supramolecular synthon rationale in the context of crystal engineering proved to be successful in designing a new series of LMWGs derived from PAM salts of a NSAID namely flufenamic acid. Out of the 10 PAM salts prepared, 9 salts were gelators of various solvents indicating that the hypothesis following which the PAM salts were designed was indeed significant. Single crystal X-ray structures as well as powder X-ray diffraction data of some of the gelators salts under various conditions further supported the hypothesis. The salts S1, S4, S5, and S7 were ambidextrous gelators producing gels with both ACS Paragon Plus Environment

Page 27 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

organic

solvent

remarkable

as

like

both

methyl

the

salicylate

solvents

are

and

pure

important

in

water the

which

is

context

of

exploiting the gels for self-drug-delivery applications via topical route. Various biological evaluations in the context of self-drugdelivery via topical route revealed that the salt S7 possessed both anti-inflammatory (PGE2 assay) and anti-cancer (PGE2 and cell migration assay) properties comparable to that of the parent drug FA and better than that of the rests. Thus, a topical formulation of S7 in methyl salicylate gel as well as its hydrogel could be useful in the context of self-drug-delivery. ACKNOWLEDGMENT P.D. thanks Department of Biotechnology (DBT), New Delhi for financial support (DBT project number: BT/01/CEIB/11/V/13). R.P. thanks CSIR, New Delhi

for

senior

research

fellowship

(CSIR

grant

number:

09/080(0876/2013-EMR-I). We thank, Parijat Biswas for capturing images in the cell imaging experiments. ASSOCIATED CONTENT Electronic Supplementary Information (ESI) available: spectra hydrogen

of

compounds,

bonding

detailed

parameters,

crystal CCDC

structures

1H

AUTHOR INFORMATION Corresponding Author * E-mail: [email protected] REFERENCES

ACS Paragon Plus Environment

13C

NMR

descriptions,

1590527-1590530.

crystallographic data in CIF See DOI: 10.1039/x0xx00000x

and

For

any

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(1)

Dastidar,

P.

Supramolecular

Gelling

Page 28 of 38

Agents:

Can

They

Be

Designed? Chem.Soc.Rev. 2008, 37, 2699–2715. DOI: 10.1039/B807346E. (2)

Weiss, R. G. The Past, Present, and Future of Molecular Gels.

What Is the Status of the Field, and Where Is It Going? J. Am. Chem. Soc. 2014, 136, 7519–7530. DOI: 10.1021/ja503363v. (3)

Jones, C. D.; Steed, J. W. Gels with Sense : Supramolecular

Materials That Respond to Heat , Light and Sound. Chem.Soc.Rev. 2016, 45, 6546–6596. DOI: 10.1039/C6CS00435K. (4)

Tomasini, C.; Castellucci, N. Peptides and peptidomimetics

that behave as low molecular weight gelators. Chem. Soc. Rev. 2013, 42, 156–172. DOI: 10.1039/C2CS35284B. (5) Suzuki,

M.;

Hanabusa,

K.

L-Lysine-based

low-molecular-weight

gelators. Chem. Soc. Rev. 2009, 38, 967–975. DOI: 10.1039/B816192E. (6)

Estroff, L. A.; Hamilton, A. D. Water Gelation by Small

Organic

Molecules.

Chem.

Rev.

2004,

104,

1201–1217.

DOI: 10.1021/cr0302049. (7)

Hirst, A. R.; Smith, D. K.; Feiters, M. C.; Geurts, H. P. M.;

Wright, A. C. Two-Component Dendritic Gels: Easily Tunable Materials. J. Am. Chem. Soc. 2003, 125, 9010–9011. DOI: 10.1021/ja036111q. (8)

Draper,

E.

R.;

Adams,

D.

J.

How

should

multicomponent

supramolecular gels be characterised ? Chem. Soc. Rev. 2018, 47, 3395– 3405. DOI: 10.1039/C7CS00804J. (9)

Cross, E. R.; Sproules, S.; Schweins, R.; Draper, E. R.;

Adams, D. J. Controlled Tuning of the Properties in Optoelectronic Self-Sorted

Gels.

J.

Am.

Chem.

Soc.

2018,

10.1021/jacs.8b05359. ACS Paragon Plus Environment

140,

8667–8670.

DOI:

Page 29 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

(10)

Babu, S. S.; Prasanthkumar, S.; Ajayaghosh, A. Self-Assembled

Gelators for Organic Electronics. Angew. Chem. Int. Ed. 2012, 51, 1766– 1776. DOI: 10.1002/anie.201106767. (11)

Ikeda, M.; Fukuda, K.; Tanida, T.; Yoshii, T.; Hamachi, I. A

supramolecular hydrogel containing boronic acid-appended receptor for fluorocolorimetric sensing of polyols with a paper platform. Chem. Commun.

2012,

48,

2716–2718.

DOI:

10.1039/C2CC17503G. (12)

Zurcher, D. M.; Adhia, Y. J.; Romero, J. D.; McNeil, A. J.

Modifying a known gelator scaffold for nitrite detection. Chem. Commun. 2014, 50, 7813–7816. DOI: 10.1039/C4CC02504K. (13)

Wynne, A.; Whitefield, M.; Dixon, A. J.; Anderson, S. An

effective, cosmetically acceptable, novel hydro-gel emollient for the management of dry skin conditions. Journal of Dermatological Treatment 2002, 13, 61–66. DOI: 10.1080/095466302317584403. (14)

Jung, J. H.; Ono, Y.; Hanabusa, K.; Shinkai, S. Creation of

Both Right-Handed and Left-Handed Silica Structures by Sol - Gel Transcription

of

Organogel

Fibers

Comprised

of

Chiral

Diaminocyclohexane Derivatives. J. Am. Chem. Soc. 2000, 122, 5008–5009. DOI: 10.1021/ja000449s. (15)

Escuder,

B.;

Rodrıguez-Llansola,

F.;

Miravet,

J.

F.

Supramolecular gels as active media for organic reactions and catalysis. New J. Chem. 2010, 34, 1044–1054. DOI: 10.1039/B9NJ00764D. (16)

Carretti, E.; Bonini, M.; Dei, L.; Berrie, B. H.; Angelova,

L. V.; Baglioni, P.; Weiss, R. G. New Frontiers in Materials Science

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 30 of 38

for Art Conservation: Responsive Gels and Beyond. Acc. Chem. Res 2010, 43, 751–760. DOI: 10.1021/ar900282h. (17)

van Bommel, K. J. C.; Stuart, M. C. A.; Feringa, B. L.; van

Esch, J. Two-Stage enzyme mediated drug release from LMWG hydrogels. Org.Biomol.Chem. 2005, 3, 2917–2920. DOI: 10.1039/B507157G. (18)

Kalafatovic, D.; Nobis, M.; Son, J.; Anderson, K. I.; Ulijn,

R. V. Biomaterials MMP-9 triggered self-assembly of doxorubicin nano fiber depots halts tumor growth. Biomaterials 2016, 98, 192–202. DOI: 10.1016/j.biomaterials.2016.04.039. (19)

Lee, K. Y.; Mooney, D. J. Hydrogels for Tissue Engineering.

Chem. Rev. 2001, 101, 1869–1880. DOI: 10.1021/cr000108x. (20) tissue

Khademhosseini, A.; Langer, R. Microengineered hydrogels for engineering.

Biomaterials

2007,

28,

5087–5092.

DOI:

10.1016/j.biomaterials.2007.07.021. (21)

Bhuniya, S.; Seo, Y. J.; Kim, B. H. (S)-(+)-Ibuprofen-based

hydrogelators: an approach toward anti-inflammatory drug delivery. Tetrahedron

Lett.

2006,

47,

7153–7156.

DOI:

10.1016/j.tetlet.2006.08.002. (22)

Vemula, P. K.; Cruikshank, G. A.; Karp, J. M.; John, G. Self-

assembled prodrugs : An enzymatically triggered drug-delivery platform. Biomaterials

2009,

30,

383–393.

DOI:

10.1016/j.biomaterials.2008.09.045. (23)

Matson, J. B.; Stupp, S. I. Drug release from hydrazone-

containing peptide amphiphiles. Chem. Commun. 2011, 7962–7964. DOI: 10.1039/C1CC12570B.

ACS Paragon Plus Environment

Page 31 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

(24)

Dastidar, P.; Roy, R.; Parveen, R.; Sarkar, K. Supramolecular

Synthon Approach in Designing Molecular Gels for Advanced Therapeutics. Adv. Therap. 2019, 2, 1800061. DOI: 10.1002/adtp.201800061. (25)

Van Esch, J. H.; Feringa, B. L. New Functional Materials

Based on Self-Assembling Organogels: From Serendipity towards Design. Angew.

Chem.

Int.

Ed.

2000,

39,

2263–2266.

DOI:

10.1002/1521-

3773(20000703)39:133.0.CO;2-V. (26)

Love, C. S.; Chechik, V.; Smith, D. K.; Brennan, C. Robust

Gels Created Using a Self-Assembly and Covalent Capture Strategy. Chem. Commun., 2005, 5647–5649. DOI: 10.1039/B511699F. (27)

Kiyonaka, S.; Sada, K.; Yoshimura, I.; Shinkai, S.; Kato, N.;

Hamachi,

I.

Semi-wet

peptide/protein

array

using

supramolecular

hydrogel. Nat. Mater. 2004, 3, 58–64. DOI: 10.1038/nmat1034. (28)

Lan, Y.; Corradini, M. G.; Weiss, R. G.; Raghavan, S. R.;

Rogers, M. A. To gel or not to gel: correlating molecular gelation with solvent

parameters.

Chem.

Soc.

Rev.

2015,

44,

6035–6058.

DOI:

10.1039/C5CS00136F. (29)

Ghosh, S.; Praveen, V. K.; Ajayaghosh, A. The Chemistry and

Applications of π-Gels. Annu. Rev. Mater. Res. 2016, 46, 235–262.DOI: 10.1146/annurev-matsci-070115-031557. (30) V.K.;

Mishra, R.K.; Das, S.; Vedhanarayanan, B.; Das, G.; Praveen, Ajayaghosh,

In Molecular Monographs

in

Gels:

A.

Stimuli-responsive

Structure

Supramolecular

and

Dynamics;

Chemistry

Supramolecular (Eds.:

Series,

R.

Royal

G.

Gels. Weiss)

Society

Chemistry, Cambridge, 2018; pp 190– 226.DOI: 10.1039/9781788013147.

ACS Paragon Plus Environment

of

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(31)

Luboradzki,

R.;

Gronwald,

O.;

Page 32 of 38

Ikeda,

M.;

Shinkai,

S.;

Reinhoudt, D. N. An attempt to predict the gelation ability of hydrogenbond-based gelators utilizing a glycoside library. Tetrahedron 2000, 56, 9595–9599. DOI: 10.1016/S0040-4020(00)00915-7. (32)

George, M.; Weiss, R. G. Molecular Organogels. Soft Matter

Comprised of Low-Molecular-Mass Organic Gelators and Organic Liquids. Acc. Chem. Res 2006, 39, 489–497. DOI: 10.1021/ar0500923. (33)

Desiraju,

G.

R.

Supramolecular

Synthons

in

Crystal

Engineering—A New Organic Synthesis. Angew. Chem. Int. Ed. Engl. 1995, 34, 2311–2327. DOI: 10.1002/anie.199523111. (34)

Desiraju, G. R.; Vittal J. J.; Ramanan A.; Crystal Engineering

– A Textbook, World Scientific, Singapore, 2011. (35) crystal

James, S. L.; Lloyd, G. O.; Zhang, J. Supramolecular gels in engineering.

CrystEngComm

2015,

17,

7976–7977.

DOI:

10.1039/C5CE90184G. (36)

Dastidar, P.; Das, U. K.; Adalder, T. K.; Majumder, J.; Roy, R.

Designing Charge-Assisted Hydrogen Bonded Supramolecular Gelators. In Hydrogen Bonded Supramolecular Materials; (Eds.: Z.-T. Li, L.-Z. Wu), Springer, Heidelberg, 2015; pp 101–131. ISBN 978-3-662-45780-1. (37)

Ballabh,

A.;

Trivedi,

D.

R.;

Dastidar,

P.

New

Series

of

Organogelators Derived from a Combinatorial Library of Primary Ammonium Monocarboxylate

Salts.

Chem.

Mater.

2006,

18,

3795–3800.

DOI:

10.1021/cm0605015. (38)

Trivedi,

D.

R.;

Ballabh,

A.;

Dastidar,

P.;

Ganguly,

B.

Structure-Property Correlation of a New Family of Organogelators Based on Organic Salts and Their Selective Gelation of Oil from Oil/Water ACS Paragon Plus Environment

Page 33 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Mixtures.

Chem.

Eur.

J.

2004,

10,

5311–5322.

DOI:

10.1002/chem.200400122. (39)

Adalder, T. K.; Dastidar, P. The Role of Secondary Ammonium

Cations in Controlling the Conformation of C 3 -Symmetric Acid Moieties and

Its

Implication

for

the

Design

of

Supramolecular

Capsules.

CrystEngComm 2014, 16, 4867–4876. DOI: 10.1039/C3CE42655F. (40)

Ballabh, A.; Trivedi, D. R.; Dastidar, P. From Nonfunctional

Lamellae to Functional Nanotubes. Org. Lett. 2006, 8, 1271–1274. DOI: 10.1021/ol053000i. (41)

Zhao, F.; Ma, M. L.; Xu, B. Molecular hydrogels of therapeutic

agents. Chem. Soc. Rev. 2009, 38, 883–891. DOI: 10.1039/B806410P. (42)

Gujrati, V.; Kim, S.; Kim, S. H.; Min, J. J.; Choy, H. E.;

Kim, S. C.; Jon, S. Bioengineered Bacterial Outer Membrane Vesicles as Cell-Specific Drug-Delivery Vehicles for Cancer Therapy. ACS Nano 2014, 8, 1525–1537. DOI: 10.1021/nn405724x. (43)

Sumer

Bolu,

B.;

Manavoglu

Gecici,

E.;

Sanyal,

R.

Combretastatin A-4 Conjugated Antiangiogenic Micellar Drug Delivery Systems Using Dendron-Polymer Conjugates. Mol. Pharmaceutics. 2016, 13, 1482–1490. DOI: (44)

10.1021/acs.molpharmaceut.5b00931.

Lukanov, B.; Firoozabadi, A. Molecular Thermodynamic Modeling

of Reverse Micelles and Water-in-Oil Microemulsions. Langmuir 2016, 32, 3100–3109. DOI: 10.1021/acs.langmuir.6b00100. (45)

Du, X.; Zhou, J.; Shi, J.; Xu, B. Supramolecular Hydrogelators

and Hydrogels: From Soft Matter to Molecular Biomaterials. Chem. Rev. 2015, 115, 13165–13307. DOI: 10.1021/acs.chemrev.5b00299.

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(46)

Page 34 of 38

Valery, C.; Maıte, P.; Robert, B.; Gulik-Krzywicki, Thaddee

Theyencheri, N.; Dedieu, J.; Keller, G.; Torres, M.; Cherif-cheikh, R.; Calvo,

P.;

Artzner,

F.

Biomimetic

Organization:

Octapeptide

Self-

Assembly into Nanotubes of Viral Capsid-like Dimension. PNAS 2003, 100, 10258–10262. DOI: 10.1073/pnas.1730609100. (47)

Xing, B.; Yu, C.-W.; Chow, K.-H.; Ho, P.-L.; Fu, D.; Xu, B.

Hydrophobic Interaction and Hydrogen Bonding Cooperatively Confer a Vancomycin Hydrogel: A Potential Candidate for Biomaterials. J. Am. Chem. Soc. 2002, 124, 14846–14847. DOI: 10.1021/ja028539f. (48)

Yang, Z.; Liang, G.; Ma, M.; Abbah, A. S.; Lu, W. W.; Xu, B.

D-Glucosamine-based supramolecular hydrogels to improve wound healing. Chem. Commun. 2007, No. 8, 843–845. DOI: 10.1039/B616563J. (49)

Parveen, R.; Sravanthi, B.; Dastidar, P. Rationally Developed

Organic Salts of Tolfenamic Acid and Its β-Alanine Derivatives for Dual Purposes as an Anti-Inflammatory Topical Gel and Anticancer Agent. Chem. Asian J. 2017, 12, 792–803. DOI: 10.1002/asia.201700049. (50) Designing

Majumder, J.; Deb, J.; Das, M. R.; Jana, S. S.; Dastidar, P. a

simple

organic

salt-based

supramolecular

topical

gel

capable of displaying in vivo self-delivery application. Chem. Commun. 2014, 50, 1671–1674. DOI: 10.1039/C3CC48513G. (51)

Raghavan, S. R.; Cipriano, B. H. Gel Formation: Phase Diagrams

Using Tabletop Rheology and Calorimetry. In Molecular Gels; Springer, 2006; pp 241–252. ISBN 978-1-4020-3689-7. (52)

Sahoo, P.; Sankolli, R.; Lee, H.; Raghavan, S. R.; Dastidar,

P. Gel Sculpture: Moldable, Load-Bearing and Self-Healing Non-Polymeric

ACS Paragon Plus Environment

Page 35 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Supramolecular Gel Derived from a Simple Organic Salt. Chem. Eur. J. 2012, 18, 8057–8063. DOI: 10.1002/chem.201200986. (53)

Yu, L.; Ding, J. Injectable Hydrogels as Unique Biomedical

Materials. Chem.Soc.Rev. 2008, 37, 1473–1481. DOI: 10.1039/b713009k. (54)

Wang, Q.-S.; Xiang, Y.; Cui, Y.-L.; Lin, K.-M.; Zhang, X.-F.

Dietary Blue Pigments Derived from Genipin, Attenuate Inflammation by Inhibiting

LPS-Induced

Inactivation.

INOS

PLoS

One

and

COX-2

Expression

2012,

7,

via

the

e34122.

NF-KB DOI:

10.1371/journal.pone.0034122. (55)

Heiss, E.; Herhaus, C.; Klimo, K.; Bartsch, H.; Gerhäuser,

C. Nuclear Factor ΚB Is a Molecular Target for Sulforaphane-Mediated Anti-Inflammatory Mechanisms. J. Biol. Chem. 2001, 276, 32008–32015. DOI: 10.1074/jbc.M104794200. (56)

Balkwill, F.; Mantovani, A. Inflammation and cancer: back to

Virchow? Lancet 2001, 357, 539–545. DOI: 10.1016/S0140-6736(00)040460. (57) Shacter E.; Weitzman S. A. Chronic inflammation and cancer. Oncology 2002, 16, 217 – 226. (58)

Basu, G. D.; Pathangey, L. B.; Tinder, T. L.; Gendler, S. J.;

Mukherjee, P. Mechanisms underlying the growth inhibitory effects of the cyclo-oxygenase-2 inhibitor celecoxib in human breast cancer cells. Breast Cancer Res. 2005, 7, R422. DOI: 10.1186/bcr1019. (59)

Ristimäki,

A.; Sivula,

A.; Lundin,

J.; Lundin,

M.; Salminen,

T.; Haglund, C.; Joensuu, H.; Isola, J. Prognostic significance of elevated

cyclooxygenase-2

expression

in

Res. 2002, 62, 632-5. ACS Paragon Plus Environment

breast

cancer.

Cancer

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 36 of 38

(60) Mayoral, R.; Fernández-Martínez, A.; Boscá, L.; Martín-Sanz, P. Prostaglandin E carcinoma

2

promotes migration and adhesion in hepatocellular

cells.

Carcinogenesis

2005,

26,

753–761.

DOI:

10.1093/carcin/bgi022. (61)

Chen,

E.

P.;

Smyth,

E.

M.

COX-2

and

PGE

2-dependent

immunomodulation in breast cancer. Prostaglandins Other Lipid Mediat. 2011, 96, 14–20. DOI: 10.1016/j.prostaglandins.2011.08.005. (62)

Timoshenko, A. V.; Xu, G.; Chakrabarti, S.; Lala, P. K.;

Chakraborty, C. Role of prostaglandin E2 receptors in migration of murine and human breast cancer cells. Exp. Cell Res. 2003, 289, 265– 274. DOI: 10.1016/S0014-4827(03)00269-6. (63)

Srinivas, S.; Feldman, D. A Phase II Trial of Calcitriol and

Naproxen in Recurrent Prostate Cancer. Anticancer Res. 2009, 29, 3605– 3610. (64) naproxen

Deb, J.; Majumder, J.; Bhattacharyya, S.; Jana, S. S. A novel derivative

capable

of

displaying

anti-cancer

and

anti-

migratory properties against human breast cancer cells. BMC Cancer 2014, 14, 567. DOI: 10.1186/1471-2407-14-567. (65)

Zhang, Y.; Kohler, N.; Zhang, M. Surface modification of

superparamagnetic uptake.

magnetite

Biomaterials

2002,

nanoparticles 23,

and

1553–1561.

9612(01)00267-8.

Table 1. Gelation table of S1-S10.

ACS Paragon Plus Environment

their DOI:

intracellular 10.1016/S0142-

Page 37 of 38 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Tgel = gel to sol dissociation temperature at MGC; Solvents

S1

S2

S3

S4

S5

S6

S7

S8

S9

S10

MGC (wt/vv) / Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)) / Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)/ Tgel

MGC (wt/vv)

Methyl salicylate

soluble

ppt

soluble

Soluble

3.7

soluble

4/50°C

gelatini ous ppt

soluble

Water

4/49°C

Soluble

soluble

4/55°C

3.7/53°C

ppt

ppt

ppt

soluble

Toluene

ppt

3/69°C

soluble

ppt

Soluble

Soluble

gelatinio us ppt

ppt

gelatinio gelatinio us ppt us ppt

gelatinio us ppt

soluble

3.7/

Soluble

3.8/65°C

gelatinio us ppt

4/53°C

gelatinio us ppt

3.7 /80°C

soluble

soluble

ppt

gelatinio us ppt

ppt

3.3/

/55°C

Nitro-bn

Chloro-bn

Bromo-bn

ppt

ppt

soluble

soluble

64°C

soluble

3/52°C

2.1/

/ Tgel ppt

gelatinio us ppt

ppt

3.7/56°C

73°C

soluble

4/60°C

ppt

61°C

gelatinio us ppt

gelatinio ppt us ppt

mesitylene

ppt

gelatinio us ppt

ppt

4/ 60°C

4/55°C

ppt

gelatinio us ppt

ppt

ppt

ppt

m-xylene

soluble

ppt

gelatinio us ppt

4/55°C

soluble

soluble

ppt

gelatini ous ppt

ppt

ppt

o-xylene

soluble

ppt

gelatinio us ppt

gelatinio us ppt

soluble

soluble

ppt

ppt

ppt

ppt

p-xylene

soluble

ppt

ppt

gelatinio us ppt

soluble

soluble

ppt

ppt

soluble

ppt

Acetonitrile

ppt

gelatino us ppt

ppt

ppt

gelatino us ppt

ppt

ppt

gelatino us ppt

ppt

soluble

DMSO

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

DMF

soluble

gelatino us ppt

soluble

soluble

soluble

soluble

Soluble

soluble

gelatino us ppt

soluble

DMA

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

soluble

ppt = precipitate; MGC = minimum gelator concentration.

For Table of Contents Use Only. ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 38 of 38

Supramolecular Gels Derived from Simple Organic Salts of Flufenamic acid: Design, Synthesis, Structures and Plausible Biomedical Application Rumana Parveen, Bandi Jayamma and Parthasarathi Dastidar*

38 ACS Paragon Plus Environment