methacrylamide Hydrochloride-Based - ACS Publications - American

Dec 1, 2017 - half-life, lack of specific cell targeting, low cellular uptake and side effect toxicity. Consequently, to deliver siRNA effectively to ...
0 downloads 4 Views 2MB Size
Subscriber access provided by READING UNIV

Article

Well-defined cationic N-[3-(dimethylamino)propyl]methacrylamide hydrochloride based (co)polymers for siRNA delivery Pratyawadee Singhsa, Diana Diaz-Dussan, Hathaikarn Manuspiya, and Ravin Narain Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.7b01475 • Publication Date (Web): 01 Dec 2017 Downloaded from http://pubs.acs.org on December 4, 2017

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 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

Biomacromolecules

1

Well-defined cationic N-[3-

2

(dimethylamino)propyl]methacrylamide

3

hydrochloride based (co)polymers for siRNA

4

delivery

5

Pratyawadee Singhsa†,‡, Diana Diaz-Dussan‡, Hathaikarn Manuspiya†,*, Ravin Narain‡,*

6



7

Materials Technology, Chulalongkorn University, Soi Chulalongkorn 12, Pathumwan, Bangkok

8

10330, Thailand.

9



The Petroleum and Petrochemical College, Center of Excellence on Petrochemical and

Department of Chemical and Materials Engineering, Donadeo Innovation Centre for

10

Engineering, 116 Street and 85 Avenue, Edmonton, AB T6G 2G6, Canada.

11

KEYWORDS: N-[3-(dimethylamino)propyl]methacrylamide, RAFT polymerization, cationic

12

glycopolymer, cytotoxicity, siRNA delivery, EGFR knockdown.

13

ACS Paragon Plus Environment

1

Biomacromolecules 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 41

1

ABSTRACT: Cationic glycopolymers have shown to be excellent candidates for the fabrication

2

of gene delivery devices due to their ability to electrostatically interact with negatively charged

3

nucleic acids and the carbohydrate residues ensure enhanced stability and low toxicity of the

4

polyplexes. The ability to engineer the polymers for optimized compositions, molecular weights

5

and architectures is critical in the design of effective gene delivery vehicles. Therefore, in this

6

study, the aqueous reversible addition-fragmentation chain transfer polymerization (RAFT) was

7

used to synthesize well-defined cationic glycopolymers with various cationic segments. For the

8

preparation of cationic parts, N-[3-(dimethylamino)propyl]methacrylamide hydrochloride

9

(DMAPMA.HCl), water-soluble methacrylamide monomer containing tertiary amine, was

10

polymerized to produce DMAPMA.HCl homopolymer which was then used as macroCTA in the

11

block copolymerization with two other methacrylamide monomers containing different pendant

12

groups, namely 2-aminoethyl methacrylamide hydrochloride (AEMA) (with primary amine) and

13

N-(3-aminopropyl) morpholine methacrylamide (MPMA) (with morpholine ring). In addition,

14

statistical copolymers of DMAPMA.HCl with either AEMA or MPMA were also synthesized.

15

All resulting cationic polymers were utilized as macroCTA for the RAFT copolymerization with

16

2-lactobionamidoethyl methacrylamide (LAEMA) which consists of the pendent galactose

17

residues to achieve DMAPMA.HCl-based glycopolymers. From the in vitro cytotoxicity study,

18

the cationic glycopolymers showed better cell viabilities than the corresponding cationic

19

homopolymers. Furthermore, complexation of the cationic polymers with siRNA, cellular uptake

20

of the resulting polyplexes and gene knockdown efficiencies were evaluated.

21

polymers/glycopolymers demonstrated good complexation ability with siRNA at low weight

22

ratios. Among these cationic polymer-siRNA polyplexes, the polyplexes prepared from the two

23

glycopolymers, P(DMAPMA65-b-LAEMA15) and P[(DMAPMA65-b-MPMA63)-b-LAEMA16],

All cationic

ACS Paragon Plus Environment

2

Page 3 of 41 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

Biomacromolecules

1

showed outstanding results in the cellular uptake, high EGFR knockdown, and low post-

2

transfection toxicity, suggesting the great potential in siRNA delivery of these novel

3

glycopolymers.

4

Introduction

5

In gene therapeutics, small interfering RNA (siRNA), small double-stranded RNA molecules

6

that silence gene expression by inducing the RNA-induced silencing complex (RISC) to cleave

7

messenger RNA (mRNA),1 has become a promising gene medicine for the specific, post-

8

transcriptional knockdown of disease-causing genes 2. siRNA has demonstrated great potential in

9

treatments of cancer, viral infections, autoimmune and neurodegenerative diseases.3-8 However,

10

the usefulness of siRNA can be limited due to their several hurdles including poor ribonuclease

11

resistance, short half-life, lack of specific cell targeting, low cellular uptake and side effect

12

toxicity. Consequently, to deliver siRNA effectively to desired target cells, novel advanced

13

delivery systems are necessary. Non-viral gene delivery systems have been greatly studied,9, 10

14

and mostly developed from cationic polymers which can electrostatically interact with negatively

15

charged siRNA to form stable complexes preventing the siRNA from enzymatic degradation and

16

destabilization by serum proteins in systemic circulation.11-14 The cationic polymers are ideal

17

candidates as siRNA carriers because of their versatility and easy manipulation required to

18

address a wide range of gene therapy applications. To improve the efficiency of siRNA delivery,

19

pH-sensitive, protonable cationic polymers have been developed, mainly due to their high

20

buffering capacity that mediate endosomal release by acting as ‘proton sponges’.15,

21

examples of cationic polymers extensively studied are polyethyleneimine (PEI),17-19 poly(amino

22

amine),14, 20, 21 and poly-L-lysine (PLL).22, 23

16

The

ACS Paragon Plus Environment

3

Biomacromolecules 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 41

1

The use of cationic polymeric carriers still has another serious obstacle in gene delivery, the

2

cationic nature of conventional polyplexes leads to severe toxicity at the cellular and systemic

3

levels including blood coagulation. For that reason, neutral hydrophilic polymers such as

4

poly(ethylene glycol) (PEG) were attached to promote the serum-tolerant function of most

5

existing polycation carriers. PEGylation of siRNA carriers represents the most exploited strategy

6

to avoid systemic toxicity of polyplexes, and increase the carrier stabilization in the

7

bloodstream.24,

8

circulating PEGylated carriers was explored to cause few immune responses,26 encouraged the

9

evaluation of different neutral hydrophilic polymers with same characteristics in term of induced

10

25

However, the accelerated blood clearance phenomenon induced by long-

stealth properties and physical carrier stabilization.

11

Natural cationic polysaccharides, for example chitosan, dextran, and β-cyclodextrin, have

12

received attention to be utilized for gene delivery, as carbohydrates are believed to be involved in

13

condensing DNA via hydrogen bonding, thus reducing the need of excess cationic charge and

14

hence decreasing the toxicity of the systems.27-30 In addition, the monomeric carbohydrate units

15

can interact with specific cell receptors, suggesting the potential of action as targeting agents of

16

polysaccharide carriers for gene delivery with no further modification needed.31, 32 Nevertheless,

17

the naturally occurring carbohydrate-based carriers have the limitation in controlling their

18

structures, so the synthesis of controllable molecular weight and architecture

19

containing carbohydrates, which so-called glycopolymers,33,

20

cationic glycopolymers that have been studied extensively in gene delivery are methacrylamide-

21

based glycopolymers. The copolymers of methacrylamide polymers containing primary amines,

22

3-aminopropyl methacrylamide (APMA) and 2-amino ethyl methacrylamide (AEMA), with

23

sugar-modified methacrylamide polymers including 3-gluconamidopropyl methacrylamide

34

polymers

is desirable. The examples of

ACS Paragon Plus Environment

4

Page 5 of 41 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

Biomacromolecules

1

(GAPMA) and 2-lactobionamidoethyl methacrylamide (LAEMA) were reported to successfully

2

formed polyplexes with plasmid DNA and provided good gene delivery efficiency in HepG2

3

cells in terms of comparable gene expression and lower toxicity, as compared to PEI.35, 36

4

N-[3-(dimethylamino)propyl]methacrylamide (DMAPMA) is a water-soluble methacrylamide

5

monomer which contains tertiary amine providing cationic polymers after polymerization.

6

DMAPMA is widely used for the synthesis of polymers in many applications, especially in

7

biomedical applications, DMAPMA has been considered towards the design of antimicrobial

8

polymers,37 temperature and/or pH sensitive polymers,38-41 protein recognition,42 protein

9

separation,43 and non-viral gene delivery systems.44-52 For the siRNA delivery purpose, the

10

DMAPMA polymer was modified with neutral hydrophilic polymers such as PEG and poly[N-

11

(2-hydroxypropyl)methacrylamide] (PHPMA) to reduce the cytotoxicity.46, 48, 52 Most of homo-

12

and copolymerizations of DMAPMA were carried out by the reversible addition-fragmentation

13

chain transfer polymerization (RAFT), a controlled/living radical polymerization (CLRP)

14

technique. The RAFT polymerization is a highly versatile technique and can provide a good

15

control over the molecular weight and molecular weight distribution for the synthesis of

16

polymers.53, 54 However, in the aqueous RAFT polymerization at neutral condition, due to the

17

presence of tertiary amines, the unprotonated form of DMAPMA can cause hydrolysis or

18

aminolysis of a chain transfer agent (CTA) such as cyanopentanoic acid dithiobenzoate (CTP),

19

resulting in a loss of chain ends and a decrease in controlled polymerization ability.55,

20

Additionally, the dithioester chain ends in the resultant polymers were degraded easily if the

21

polymers were purified by dialysis against pure deionized water.46, 57 Therefore, when synthesize

22

DMAPMA polymers, the acidic aqueous medium is required for the RAFT polymerization and

23

purification.

56

ACS Paragon Plus Environment

5

Biomacromolecules 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 6 of 41

1

From the mentioned problems associated with the unprotonated DMAPMA, DMAPMA

2

hydrochloride (DMAPMA.HCl) which is protonated by hydrochloric acid before subjecting to

3

polymerization, was developed and studied for its ability in homo- and copolymerization by

4

RAFT in the previous work.58 The well-controlled homopolymers of DMAPMA.HCl were

5

obtained by the RAFT polymerization in acidic medium and simple purification with

6

precipitation in acetone which allowed the good retention of dithioester chain end groups.

7

Consequently, the p(DMAPMA.HCl)-based macroCTA was prepared and successfully used in

8

the diblock copolymerization with other monomers. Moreover, the well-defined statistical

9

copolymers of DMAPMA.HCl were attained as well.

10

Although DMAPMA has been popularly used in RAFT copolymerizations, it was mostly

11

subjected as a second-block or third-block co-monomer in chain-extension of block

12

copolymerizations, and there was no report of utilizing DMAPMA in synthesis of glycopolymers

13

for gene delivery applications. Therefore, the aim of this work was to prepare well-defined

14

cationic DMAPMA.HCl-based polymers/glycopolymers with different architectures by using

15

P(DMAPMA.HCl)-based macroCTA via the aqueous RAFT polymerization. The co-monomers

16

used were methacrylamide monomers containing various pendant groups; 2-aminoethyl

17

methacrylamide hydrochloride (AEMA) (with

18

methacrylamide

19

methacrylamide (MPMA) (with morpholine). Then, the resulting polymers/glycopolymers

20

cytotoxicities were evaluated in HeLa (cervical) cancer cells by the tetrazolium MTT

21

colorimetric assay. For the siRNA delivery purpose, the cationic polymer/glycopolymer-siRNA

22

complexation, cellular uptake of the resulting polyplexes, transfection efficiencies, epidermal

23

growth factor receptor (EGFR) siRNA knockdown, and post-transfection toxicity were studied.

(LAEMA)

(with

saccharide),

primary amine), 2-lactobionamidoethyl and

N-(3-aminopropyl)

morpholine

ACS Paragon Plus Environment

6

Page 7 of 41 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

Biomacromolecules

Experimental section

2

Materials. N-[3-(dimethylamino)propyl]methacrylamide (DMAPMA) was purchased from

3

Sigma-Aldrich Chemicals, and protonated with hydrochloric acid as ascribed in previous work.58

4

N-(3-aminopropyl) morpholine methacrylamide (MPMA), 2-aminoethyl methacrylamide

5

hydrochloride (AEMA), and 2-lactobionamidoethyl methacrylamide (LAEMA) were synthesized

6

according to previous reports.58-61 The CTA, cyanopentanoic acid dithiobenzoate (CTP), was

7

synthesized as previously described.62

8

hydrochloric acid (ACS reagent, 37%) were purchased from Sigma Aldrich Chemicals and used

9

as received. Methanol, 2-propanol, tetrahydrofuran (THF), N,N’-dimethyl formaldehyde (DMF),

10

dimethylsulfoxide (DMSO), and acetone were purchased from Caledon Laboratory Chemicals.

11

Milli-Q water was used for all the synthesis experiments. Dulbecco's Modified Eagle Medium

12

(DMEM), Opti-MEM (OMEM), 0.25% trypsin−EDTA, fetal bovine serum (FBS) and

13

Streptomycin (5000 µg/mL)-Penicillin (5000 U/mL) were obtained from Gibco. Lipofectamine

14

was purchased from Invitrogen Control. EGFR siRNA−FITC conjugate, human EGFR-specific

15

small interfering RNA (EGFR siRNA), primary antibody (rabbit polyclonal EGFR specific IgG)

16

and FITC-conjugated control siRNA were purchased from Santa Cruz Biotechnology.

4,4’-Azobis(4-cyanovaleric acid) (ACVA) and

17

Synthesis of the DMAPMA.HCl-macro chain transfer agent (CTA). The polymerization of

18

DMAPMA.HCl was conducted using RAFT technique at 70 °C, ACVA as the initiator and CTP

19

as the CTA. In a typical protocol, in a 10 mL Schlenk tube, DMAPMA.HCl (1.24 g, 6 mmol),

20

CTP (0.017 g, 0.06 mmol, target DPn = 100) and ACVA (0.008 g, 0.03 mmol) were dissolved in

21

6 mL of a solvent mixture of acidic water (1:50 v/v mixture of hydrochloric acid and water, pH
90% cell viability), which was better than their corresponding free polymers, suggesting that

2

the presence of nucleic acid molecules could mask part of the cytotoxic effect of the polymers.44

3

This

4

polymers/glycopolymers supported the benefits of utilizing these systems in siRNA delivery.

high

biocompatibility

of

the

polyplexes

prepared

by

DMAPMA.HCl-based

5

Figure 6. Post-transfection toxicity of the cationic polymer-siRNA polyplexes in HeLa cells. Cell viability was assessed by Janus Green assay 48 h post-transfection. 6

Conclusion

7

In this work, with aqueous RAFT polymerization, the DMAPMA.HCl homopolymer (P1)

8

was synthesized and utilized as P(DMAPMA.HCl)-based macroCTA in chain extension with

9

either AEMA or MPMA to produce well-defined diblock copolymers (P3 and P4). In addition,

10

for investigating the effect of polymer architectures, the well-controlled statistical copolymers of

11

DMAPMA.HCl with AEMA or MPMA (P7 and P8) were also prepared. Then, DMAPMA.HCl-

12

based glycopolymers (P2, P5, P6, P9, and P10) were successfully produced by using each of the

13

obtained polymers as the macroCTA in the block copolymerization with the same amounts of

14

LAEMA. All resulting polymers were evaluated for their cytotoxicity in HeLa cells and, as

ACS Paragon Plus Environment

33

Biomacromolecules 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 34 of 41

1

compared to the DMAPMA.HCl homopolymer, the copolymers with MPMA showed lower cell

2

toxicity. On the contrary, increased cell toxicities were found in the copolymers with AEMA.

3

Interestingly, all DMAPMA.HCl-based glycopolymers demonstrated significantly improved

4

biocompatibility. Consequently, the resulting cationic polymers/glycopolymers were determined

5

their ability of complexation with siRNA, and the results showed that they could form stable

6

polyplexes at low weight ratios. However, the copolymers with AEMA (P3 and P7) and their

7

corresponding glycopolymers (P5 and P9) were excluded from further studies due to their high

8

toxicity. The other polymers were evaluated for their potential of siRNA delivery, the

9

hydrodynamic sizes of the resulting polyplexes were in range of 350-600 nm with the zeta

10

potential values about 2-6 mV, and all polyplexes were internalized to the cytoplasm of HeLa

11

cells which were visualized by the confocal fluorescence microscopy. From the transfection

12

efficiency study (EGFR knockdown), the P6 which was the glycopolymer consisting of

13

DMAPMA.HCl-MPMA block copolymer exhibited the highest EGFR silencing (~10% gene

14

expression), while the P2 (simplest DMAPMA.HCl glycopolymer) showed the higher EGFR

15

down-regulation (~30% gene expression) without off-target knockdown. Additionally, the post-

16

transfection toxicity revealed that the cationic polymer-siRNA polyplexes were highly

17

biocompatible (>90% cell viability). Therefore, the DMAPMA.HCl-based glycopolymer

18

with/without MPMA blocks can be promising materials for siRNA delivery.

19 20

ASSOCIATED CONTENT

21

Supporting Information.

22

The following files are available free of charge.

ACS Paragon Plus Environment

34

Page 35 of 41 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

Biomacromolecules

1

GPC traces and 1H NMR spectra of DMAPMA.HCl polymers/glycopolymers, and agarose gel

2

electrophoresis retardation assay of the cationic polymer-siRNA polyplexes. (PDF)

3

AUTHOR INFORMATION

4

Corresponding Authors

5

Professor Ravin Narain

6

*E-mail: [email protected]. Phone: +17804921736

7

Professor Hathaikarn Manuspiya

8

*

9

E-mail: [email protected]

ORCID

10

Pratyawadee Singhsa: 0000-0001-5492-6245

11

Diana Diaz-Dussan: 0000-0003-1778-3964

12

Ravin Narain: 0000-0003-0947-9719

13

Notes

14

The authors declare no competing financial interest.

15

ACKNOWLEDGMENT

16

This research was supported by the Natural Sciences and Engineering Council of Canada

17

(NSERC) and by the Petroleum and Petrochemical College, Chulalongkorn University, and

18

funded by the Doctoral Degree Chulalongkorn University 100th Year Birthday Anniversary

19

Scholarship.

20

REFERENCES

ACS Paragon Plus Environment

35

Biomacromolecules 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 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

Page 36 of 41

1. Meister, G.; Tuschl, T., Mechanisms of gene silencing by double-stranded RNA. Nature 2004, 431, (7006), 343-349. 2. Reischl, D.; Zimmer, A., Drug delivery of siRNA therapeutics: potentials and limits of nanosystems. Nanomed.: Nanotechnol. Biol. Med. 2009, 5, (1), 8-20. 3. Novina, C. D.; Murray, M. F.; Dykxhoorn, D. M.; Beresford, P. J.; Riess, J.; Lee, S.-K.; Collman, R. G.; Lieberman, J.; Shankar, P.; Sharp, P. A., siRNA-directed inhibition of HIV-1 infection. Nat. Med. 2003, 9, (11), 1433-1433. 4. Grimm, D., Small silencing RNAs: State-of-the-art. Adv. Drug Delivery Rev. 2009, 61, (9), 672-703. 5. Devi, G. R., siRNA-based approaches in cancer therapy. Cancer Gene Ther. 2006, 13, (9), 819-829. 6. Takeshita, F.; Ochiya, T., Therapeutic potential of RNA interference against cancer. Cancer Sci. 2006, 97, (8), 689-696. 7. Kim, H. J.; Kim, A.; Miyata, K.; Kataoka, K., Recent progress in development of siRNA delivery vehicles for cancer therapy. Adv. Drug Delivery Rev. 2016, 104, 61-77. 8. Harper, S. Q.; Staber, P. D.; He, X.; Eliason, S. L.; Martins, I. H.; Mao, Q.; Yang, L.; Kotin, R. M.; Paulson, H. L.; Davidson, B. L., RNA interference improves motor and neuropathological abnormalities in a Huntington's disease mouse model. Proc. Natl. Acad. Sci. U. S. A. 2005, 102, (16), 5820-5825. 9. Pun, S. H.; Davis, M. E., Development of a Nonviral Gene Delivery Vehicle for Systemic Application. Bioconjugate Chem. 2002, 13, (3), 630-639. 10. Foldvari, M.; Chen, D. W.; Nafissi, N.; Calderon, D.; Narsineni, L.; Rafiee, A., Non-viral gene therapy: Gains and challenges of non-invasive administration methods. J. Controlled Release 2016, 240, 165-190. 11. Aied, A.; Greiser, U.; Pandit, A.; Wang, W., Polymer gene delivery: overcoming the obstacles. Drug Discovery Today 2013, 18, (21–22), 1090-1098. 12. Gary, D. J.; Min, J. B.; Kim, Y.; Park, K.; Won, Y.-Y., The Effect of N/P Ratio on the In Vitro and In Vivo Interaction Properties of PEGylated Poly(2-(dimethylamino)ethyl methacrylate)-Based siRNA Complexes. Macromol. biosci. 2013, 13, (8), 1059-1071. 13. von Harpe, A.; Petersen, H.; Li, Y.; Kissel, T., Characterization of commercially available and synthesized polyethylenimines for gene delivery. J. Controlled Release 2000, 69, (2), 309-322. 14. Van der Aa, L. J.; Vader, P.; Storm, G.; Schiffelers, R. M.; Engbersen, J. F. J., Optimization of poly(amido amine)s as vectors for siRNA delivery. J. Controlled Release 2011, 150, (2), 177-186. 15. Akinc, A.; Thomas, M.; Klibanov, A. M.; Langer, R., Exploring polyethyleniminemediated DNA transfection and the proton sponge hypothesis. J. Gene Med. 2005, 7, (5), 657663. 16. Guidry, E. N.; Farand, J.; Soheili, A.; Parish, C. A.; Kevin, N. J.; Pipik, B.; Calati, K. B.; Ikemoto, N.; Waldman, J. H.; Latham, A. H.; Howell, B. J.; Leone, A.; Garbaccio, R. M.; Barrett, S. E.; Parmar, R. G.; Truong, Q. T.; Mao, B.; Davies, I. W.; Colletti, S. L.; SeppLorenzino, L., Improving the In Vivo Therapeutic Index of siRNA Polymer Conjugates through Increasing pH Responsiveness. Bioconjugate Chem. 2014, 25, (2), 296-307. 17. Günther, M.; Lipka, J.; Malek, A.; Gutsch, D.; Kreyling, W.; Aigner, A., Polyethylenimines for RNAi-mediated gene targeting in vivo and siRNA delivery to the lung. Eur. J. Pharm. Biopharm. 2011, 77, (3), 438-449.

ACS Paragon Plus Environment

36

Page 37 of 41 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 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

Biomacromolecules

18. Malek, A.; Merkel, O.; Fink, L.; Czubayko, F.; Kissel, T.; Aigner, A., In vivo pharmacokinetics, tissue distribution and underlying mechanisms of various PEI(–PEG)/siRNA complexes. Toxicol. Appl. Pharm. 2009, 236, (1), 97-108. 19. Werth, S.; Urban-Klein, B.; Dai, L.; Höbel, S.; Grzelinski, M.; Bakowsky, U.; Czubayko, F.; Aigner, A., A low molecular weight fraction of polyethylenimine (PEI) displays increased transfection efficiency of DNA and siRNA in fresh or lyophilized complexes. J. Controlled Release 2006, 112, (2), 257-270. 20. Patil, M. L.; Zhang, M.; Betigeri, S.; Taratula, O.; He, H.; Minko, T., Surface-Modified and Internally Cationic Polyamidoamine Dendrimers for Efficient siRNA Delivery. Bioconjugate Chem. 2008, 19, (7), 1396-1403. 21. Zhou, J.; Wu, J.; Hafdi, N.; Behr, J.-P.; Erbacher, P.; Peng, L., PAMAM dendrimers for efficient siRNA delivery and potent gene silencing. Chem. Commun. 2006, (22), 2362-2364. 22. Buyens, K.; Meyer, M.; Wagner, E.; Demeester, J.; De Smedt, S. C.; Sanders, N. N., Monitoring the disassembly of siRNA polyplexes in serum is crucial for predicting their biological efficacy. J. Controlled Release 2010, 141, (1), 38-41. 23. Hayashi, K.; Chaya, H.; Fukushima, S.; Watanabe, S.; Takemoto, H.; Osada, K.; Nishiyama, N.; Miyata, K.; Kataoka, K., Influence of RNA Strand Rigidity on Polyion Complex Formation with Block Catiomers. Macromol. Rapid Commun. 2016, 37, (6), 486-493. 24. Mao, S.; Neu, M.; Germershaus, O.; Merkel, O.; Sitterberg, J.; Bakowsky, U.; Kissel, T., Influence of Polyethylene Glycol Chain Length on the Physicochemical and Biological Properties of Poly(ethylene imine)-graft-Poly(ethylene glycol) Block Copolymer/SiRNA Polyplexes. Bioconjugate Chem. 2006, 17, (5), 1209-1218. 25. Qi, R.; Liu, S.; Chen, J.; Xiao, H.; Yan, L.; Huang, Y.; Jing, X., Biodegradable copolymers with identical cationic segments and their performance in siRNA delivery. J. Controlled Release 2012, 159, (2), 251-260. 26. Merkel, O. M.; Librizzi, D.; Pfestroff, A.; Schurrat, T.; Buyens, K.; Sanders, N. N.; De Smedt, S. C.; Béhé, M.; Kissel, T., Stability of siRNA polyplexes from poly(ethylenimine) and poly(ethylenimine)-g-poly(ethylene glycol) under in vivo conditions: Effects on pharmacokinetics and biodistribution measured by Fluorescence Fluctuation Spectroscopy and Single Photon Emission Computed Tomography (SPECT) imaging. J. Controlled Release 2009, 138, (2), 148-159. 27. Azzam, T.; Raskin, A.; Makovitzki, A.; Brem, H.; Vierling, P.; Lineal, M.; Domb, A. J., Cationic Polysaccharides for Gene Delivery. Macromolecules 2002, 35, (27), 9947-9953. 28. Strand, S. P.; Issa, M. M.; Christensen, B. E.; Vårum, K. M.; Artursson, P., Tailoring of Chitosans for Gene Delivery: Novel Self-Branched Glycosylated Chitosan Oligomers with Improved Functional Properties. Biomacromolecules 2008, 9, (11), 3268-3276. 29. Popielarski, S. R.; Mishra, S.; Davis, M. E., Structural Effects of CarbohydrateContaining Polycations on Gene Delivery. 3. Cyclodextrin Type and Functionalization. Bioconjugate Chem. 2003, 14, (3), 672-678. 30. Wang, Z. H.; Li, W. B.; Ma, J.; Tang, G. P.; Yang, W. T.; Xu, F. J., Functionalized Nonionic Dextran Backbones by Atom Transfer Radical Polymerization for Efficient Gene Delivery. Macromolecules 2011, 44, (2), 230-239. 31. Anderson, K.; Fernandez, C.; Rice, K. G., N-Glycan Targeted Gene Delivery to the Dendritic Cell SIGN Receptor. Bioconjugate Chem. 2010, 21, (8), 1479-1485. 32. Cavallaro, G.; Farra, R.; Craparo, E. F.; Sardo, C.; Porsio, B.; Giammona, G.; Perrone, F.; Grassi, M.; Pozzato, G.; Grassi, G.; Dapas, B., Galactosylated polyaspartamide copolymers

ACS Paragon Plus Environment

37

Biomacromolecules 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 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

Page 38 of 41

for siRNA targeted delivery to hepatocellular carcinoma cells. Inter. J. Pharm. 2017, 525, (2), 397-406. 33. Pearson, S.; Chen, G.; Stenzel, M. H., Synthesis of Glycopolymers. In Engineered Carbohydrate-Based Materials for Biomedical Applications, John Wiley & Sons, Inc.: 2011; pp 1-118. 34. Yilmaz, G.; Becer, C. R., Glycopolymer Code based on Well-defined Glycopolymers or Glyconanomaterials and Their Biomolecular Recognition. Frontiers in Bioengineering and Biotechnology 2014, 2. 35. Ahmed, M.; Narain, R., The effect of polymer architecture, composition, and molecular weight on the properties of glycopolymer-based non-viral gene delivery systems. Biomaterials 2011, 32, (22), 5279-5290. 36. Ahmed, M.; Jawanda, M.; Ishihara, K.; Narain, R., Impact of the nature, size and chain topologies of carbohydrate–phosphorylcholine polymeric gene delivery systems. Biomaterials 2012, 33, (31), 7858-7870. 37. Paslay, L. C.; Abel, B. A.; Brown, T. D.; Koul, V.; Choudhary, V.; McCormick, C. L.; Morgan, S. E., Antimicrobial Poly(methacrylamide) Derivatives Prepared via Aqueous RAFT Polymerization Exhibit Biocidal Efficiency Dependent upon Cation Structure. Biomacromolecules 2012, 13, (8), 2472-2482. 38. Çaykara, T.; Demiray, M.; Güven, O., Effect of type and concentration of surfactants on swelling behavior of poly[N-[3-(dimethylamino)propyl]methacrylamide-coN,Nmethylenebis(acrylamide)] hydrogels. Colloid Polym. Sci. 2005, 284, (3), 258-265. 39. Das, A.; Ray, A. R., Synthesis and characterization of poly(acrylic acid-co-N-[3(dimethylamino)propyl]-methacrylamide) hydrogel membranes for biomedical applications. J. Appl. Polym. Sci. 2008, 108, (2), 1273-1280. 40. Mishra, R. K.; Ray, A. R., Synthesis and characterization of poly{N-[3-(dimethylamino) propyl] methacrylamide-co-itaconic acid} hydrogels for drug delivery. J. Appl. Polym. Sci. 2011, 119, (6), 3199-3206. 41. Das, A.; Ghosh, S.; Ray, A. R., Unveiling the self-assembly behavior of copolymers of AAc and DMAPMA in situ to form smart hydrogels displaying nanogels-within-macrogel hierarchical morphology. Polymer 2011, 52, (17), 3800-3810. 42. Huang, J.-T.; Zhang, J.; Zhang, J.-Q.; Zheng, S.-H., Template imprinting amphoteric polymer for the recognition of proteins. J. Appl. Polym. Sci. 2005, 95, (2), 358-361. 43. Tuncel, A.; Demirgöz, D.; Patir, S.; Piskin, E., A novel approach for albumin determination in aqueous media by using temperature- and pH-sensitive N-isopropylacrylamideco-N-[3-(dimethylamino)-propyl]methacrylamide random copolymers. J. Appl. Polym. Sci. 2002, 84, (11), 2060-2071. 44. van de Wetering, P.; Moret, E. E.; Schuurmans-Nieuwenbroek, N. M. E.; van Steenbergen, M. J.; Hennink, W. E., Structure−Activity Relationships of Water-Soluble Cationic Methacrylate/Methacrylamide Polymers for Nonviral Gene Delivery. Bioconjugate Chem. 1999, 10, (4), 589-597. 45. Ahmed, M.; Narain, R., Progress of RAFT based polymers in gene delivery. Prog. Polym. Sci. 2013, 38, (5), 767-790. 46. Scales, C. W.; Huang, F.; Li, N.; Vasilieva, Y. A.; Ray, J.; Convertine, A. J.; McCormick, C. L., Corona-Stabilized Interpolyelectrolyte Complexes of SiRNA with Nonimmunogenic, Hydrophilic/Cationic Block Copolymers Prepared by Aqueous RAFT Polymerization. Macromolecules 2006, 39, (20), 6871-6881.

ACS Paragon Plus Environment

38

Page 39 of 41 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 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

Biomacromolecules

47. Zhu, J.-L.; Cheng, H.; Jin, Y.; Cheng, S.-X.; Zhang, X.-Z.; Zhuo, R.-X., Novel polycationic micelles for drug delivery and gene transfer. J. Mater. Chem. 2008, 18, (37), 44334441. 48. York, A. W.; Zhang, Y.; Holley, A. C.; Guo, Y.; Huang, F.; McCormick, C. L., Facile Synthesis of Multivalent Folate-Block Copolymer Conjugates via Aqueous RAFT Polymerization: Targeted Delivery of siRNA and Subsequent Gene Suppression. Biomacromolecules 2009, 10, (4), 936-943. 49. Kirkland-York, S.; Zhang, Y.; Smith, A. E.; York, A. W.; Huang, F.; McCormick, C. L., Tailored Design of Au Nanoparticle-siRNA Carriers Utilizing Reversible Addition−Fragmentation Chain Transfer Polymers. Biomacromolecules 2010, 11, (4), 10521059. 50. Holley, A. C.; Parsons, K. H.; Wan, W.; Lyons, D. F.; Bishop, G. R.; Correia, J. J.; Huang, F.; McCormick, C. L., Block ionomer complexes consisting of siRNA and aRAFTsynthesized hydrophilic-block-cationic copolymers: the influence of cationic block length on gene suppression. Polym. Chem. 2014, 5, (24), 6967-6976. 51. Xu, M.; Qian, J.; Suo, A.; Cui, N.; Yao, Y.; Xu, W.; Liu, T.; Wang, H., Co-delivery of doxorubicin and P-glycoprotein siRNA by multifunctional triblock copolymers for enhanced anticancer efficacy in breast cancer cells. J. Mater. Chem. B 2015, 3, (10), 2215-2228. 52. Xu, M.; Qian, J.; Suo, A.; Xu, W.; Liu, R.; Wang, H., Stimuli-responsive terpolymer mPEG-b-PDMAPMA-b-PAH mediated co-delivery of adriamycin and siRNA to enhance anticancer efficacy. RSC Adv. 2015, 5, (27), 20890-20899. 53. Chiefari, J.; Chong, Y. K.; Ercole, F.; Krstina, J.; Jeffery, J.; Le, T. P. T.; Mayadunne, R. T. A.; Meijs, G. F.; Moad, C. L.; Moad, G.; Rizzardo, E.; Thang, S. H., Living Free-Radical Polymerization by Reversible Addition−Fragmentation Chain Transfer:  The RAFT Process. Macromolecules 1998, 31, (16), 5559-5562. 54. Hinton, T. M.; Guerrero-Sanchez, C.; Graham, J. E.; Le, T.; Muir, B. W.; Shi, S.; Tizard, M. L. V.; Gunatillake, P. A.; McLean, K. M.; Thang, S. H., The effect of RAFT-derived cationic block copolymer structure on gene silencing efficiency. Biomaterials 2012, 33, (30), 7631-7642. 55. McCormick, C. L.; Lowe, A. B., Aqueous RAFT Polymerization:  Recent Developments in Synthesis of Functional Water-Soluble (Co)polymers with Controlled Structures. Acc. Chem. Res. 2004, 37, (5), 312-325. 56. McCormick, C. L.; Kirkland, S. E.; York, A. W., Synthetic Routes to Stimuli‐ Responsive Micelles, Vesicles, and Surfaces via Controlled/Living Radical Polymerization. J. Macromol. Sc., Part C 2006, 46, (4), 421-443. 57. Vasilieva, Y. A.; Thomas, D. B.; Scales, C. W.; McCormick, C. L., Direct Controlled Polymerization of a Cationic Methacrylamido Monomer in Aqueous Media via the RAFT Process. Macromolecules 2004, 37, (8), 2728-2737. 58. Singhsa, P.; Manuspiya, H.; Narain, R., Study of the RAFT homopolymerization and copolymerization of N-[3-(dimethylamino)propyl]methacrylamide hydrochloride and evaluation of the cytotoxicity of the resulting homo- and copolymers. Polym. Chem. 2017, 8, (28), 41404151. 59. Deng, Z.; Bouchékif, H.; Babooram, K.; Housni, A.; Choytun, N.; Narain, R., Facile synthesis of controlled-structure primary amine-based methacrylamide polymers via the reversible addition-fragmentation chain transfer process. J. Polym. Sci., Part A: Polym. Chem. 2008, 46, (15), 4984-4996.

ACS Paragon Plus Environment

39

Biomacromolecules 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 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

Page 40 of 41

60. Deng, Z.; Ahmed, M.; Narain, R., Novel well-defined glycopolymers synthesized via the reversible addition fragmentation chain transfer process in aqueous media. J. Polym. Sci., Part A: Polym. Chem. 2009, 47, (2), 614-627. 61. Deng, Z.; Li, S.; Jiang, X.; Narain, R., Well-Defined Galactose-Containing MultiFunctional Copolymers and Glyconanoparticles for Biomolecular Recognition Processes. Macromolecules 2009, 42, (17), 6393-6405. 62. Mitsukami, Y.; Donovan, M. S.; Lowe, A. B.; McCormick, C. L., Water-Soluble Polymers. 81. Direct Synthesis of Hydrophilic Styrenic-Based Homopolymers and Block Copolymers in Aqueous Solution via RAFT. Macromolecules 2001, 34, (7), 2248-2256. 63. Vana, P.; Davis, T. P.; Barner-Kowollik, C., Kinetic Analysis of Reversible Addition Fragmentation Chain Transfer (RAFT) Polymerizations: Conditions forInhibition, Retardation, and Optimum LivingPolymerization. Macromo. Theory Simul. 2002, 11, (8), 823-835. 64. Fischer, D.; Li, Y.; Ahlemeyer, B.; Krieglstein, J.; Kissel, T., In vitro cytotoxicity testing of polycations: influence of polymer structure on cell viability and hemolysis. Biomaterials 2003, 24, (7), 1121-1131. 65. Moghimi, S. M.; Symonds, P.; Murray, J. C.; Hunter, A. C.; Debska, G.; Szewczyk, A., A two-stage poly(ethylenimine)-mediated cytotoxicity: implications for gene transfer/therapy. Molecul.Ther. 2005, 11, (6), 990-995. 66. Cai, J.; Yue, Y.; Rui, D.; Zhang, Y.; Liu, S.; Wu, C., Effect of Chain Length on Cytotoxicity and Endocytosis of Cationic Polymers. Macromolecules 2011, 44, (7), 2050-2057. 67. Sgouras, D.; Duncan, R., Methods for the evaluation of biocompatibility of soluble synthetic polymers which have potential for biomedical use: 1 — Use of the tetrazolium-based colorimetric assay (MTT) as a preliminary screen for evaluation ofin vitro cytotoxicity. J. Mater. Sci.: Mater. Med. 1990, 1, (2), 61-68. 68. Choksakulnimitr, S.; Masuda, S.; Tokuda, H.; Takakura, Y.; Hashida, M., In vitro cytotoxicity of macromolecules in different cell culture systems. J. Controlled Release 1995, 34, (3), 233-241. 69. Chanana, M.; Gliozzi, A.; Diaspro, A.; Chodnevskaja, I.; Huewel, S.; Moskalenko, V.; Ulrichs, K.; Galla, H.-J.; Krol, S., Interaction of Polyelectrolytes and Their Composites with Living Cells. Nano Lett. 2005, 5, (12), 2605-2612. 70. Zhi, D.; Zhang, S.; Cui, S.; Zhao, Y.; Wang, Y.; Zhao, D., The Headgroup Evolution of Cationic Lipids for Gene Delivery. Bioconjugate Chem. 2013, 24, (4), 487-519. 71. Velasco, D.; Elvira, C.; San Román, J., New stimuli-responsive polymers derived from morpholine and pyrrolidine. J. Mater. Sci.: Mater. Med. 2007, 19, (4), 1453-1458. 72. Lessard, B. H.; Savelyeva, X.; Marić, M., Smart morpholine-functional statistical copolymers synthesized by nitroxide mediated polymerization. Polymer 2012, 53, (25), 56495656. 73. Piestrzeniewicz, M. K.; Wilmańska, D.; Szemraj, J.; Studzian, K.; Gniazdowski, M., Interactions of novel morpholine and hexamethylene derivatives of anthracycline antibiotics with DNA. Zeitschrift für Naturforschung C 2004, 59, (9-10), 739-748. 74. Diaz-Dussan, D.; Nakagawa, Y.; Peng, Y.-Y.; C, L. V. S.; Ebara, M.; Kumar, P.; Narain, R., Effective and Specific Gene Silencing of Epidermal Growth Factor Receptors Mediated by Conjugated Oxaborole and Galactose-Based Polymers. ACS Macro Lett 2017, 6, (7), 768-774. 75. Nguyen, J.; Szoka, F. C., Nucleic Acid Delivery: The Missing Pieces of the Puzzle? Acc. Chem. Res. 2012, 45, (7), 1153-1162.

ACS Paragon Plus Environment

40

Page 41 of 41 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 2 3 4 5 6

Biomacromolecules

76. Satpathy, M.; Mezencev, R.; Wang, L.; McDonald, J. F., Targeted in vivo delivery of EGFR siRNA inhibits ovarian cancer growth and enhances drug sensitivity. Sci. Rep. 2016, 6, 36518. 77. Dickerson, E. B.; Blackburn, W. H.; Smith, M. H.; Kapa, L. B.; Lyon, L. A.; McDonald, J. F., Chemosensitization of cancer cells by siRNA using targeted nanogel delivery. BMC Cancer 2010, 10, 10-10.

7 8 9

Table of content

10

ACS Paragon Plus Environment

41