Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

Subscriber access provided by READING UNIV

Article
Systemic mRNA Delivery to the Lungs by Functional Polyester-based Carriers
Yunfeng Yan, Hu Xiong, Xinyi Zhang, QIANG CHENG, and Daniel John Siegwart
Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.7b01356 • Publication Date (Web): 15 Nov 2017
Downloaded from http://pubs.acs.org on November 15, 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 24 Biomacromolecules

1
2
3
4 Systemic mRNA Delivery to the Lungs by Functional Polyester-based Carriers
5
6 Yunfeng Yan a,b,*, Hu Xiong b, Xinyi Zhang b, Qiang Cheng b, and Daniel J. Siegwart b,*
7
8
a
9 College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou,
10 Zhejiang 310014, China
11
12 b
Simmons Comprehensive Cancer Center, Department of Biochemistry, The University of Texas
13
Southwestern Medical Center, Dallas, TX 75390, United States
14
15 *
Corresponding authors. Email address: Daniel.Siegwart@UTSouthwestern.edu (D.J. Siegwart),
16
17 yfyan@zjut.edu.cn (Y. Yan).
18
19
20
21 Abstract
22
23
24 Messenger RNA (mRNA) has recently come into focus as an emerging therapeutic class with great
25
26 potential for protein replacement therapy, cancer immunotherapy, regenerative medicine, vaccines, and
27
28
gene editing. However, the lack of effective and safe delivery methods impedes the broad application of
29 mRNA-based therapeutics. Here, we report a robust approach to develop efficient polymeric delivery
30
31 carriers for mRNA. Lead polyesters were identified by in vitro screening of a 480-member
32
33 combinatorially modified poly(trimethylolpropane allyl ether-co-suberoyl chloride) library for the
34
35 delivery of luciferase encoding mRNA (Luc mRNA) to IGROV1 cells. Formulation of mRNA polyplex
36 nanoparticles (NPs) with Pluronic F127 decreased the surface charge. Although this improved the
37
38 stability of mRNA nanoparticles, the delivery potency decreased with increased F127 content. Thus, we
39
40 determined that NP stabilization with 5% F127 could balance the protective effects and delivery potency.
41
42 5% F127 formulated PE4K-A17-0.33C12 mRNA NPs enabled luciferase expression predominantly in
43
the lungs after intravenous injection into mice. The efficient mRNA delivery specifically to lungs by
44
45 degradable carriers suggests potential for the treatment of pulmonary diseases.
46
47
48 Keywords: Polyesters, mRNA, nanoparticles, drug delivery
49
50
51
52
53 Introduction
54
55
56 Messenger RNA (mRNA)-based therapeutics have recently generated great attention because of the
57
58
broad range of potential applications.1-3 Synthetically modified and codon-optimized mRNAs are
59
60 1
ACS Paragon Plus Environment
Biomacromolecules Page 2 of 24

1
2
3
capable of expressing encoded proteins in cells, which enables wide utilization in protein replacement
4
5 therapy, immunotherapy, regenerative medicine, and infectious disease vaccines.4-12 In particular, the
6
7 effective and controllable expression of target proteins by delivered mRNA provides a reliable approach
8
9 for the production of the key nuclease for the emerging genome editing technologies, e.g. zinc finger
10
11
nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly
12 interspaced short palindromic repeat (CRISPR)/Cas.13-18 Due to the great potential in clinical
13
14 applications, there has been significant investment in a wide array of biotechnology enterprises.1-3
15
16
17 mRNA-based therapeutics hold some conceptual advantages over other gene therapy methods.
18
19
For example, mRNA does not integrate into the host genome. Thus, there is limited risk of mutagenesis
20 or persistent expression of gene editing related proteins. mRNA is translated to functional proteins in
21
22 cytoplasm and does not need to enter the nucleus, which aids utility in non-dividing or slowly dividing
23
24 cells, e.g. dendritic cells.1, 19 Furthermore, comparing with recombinant proteins, the chemical synthesis
25
26 of mRNA is easier, cheaper, and carries a lower biosafety risk.1, 20
27
28 There are some ongoing challenges to realize the broad potential of mRNA-based therapeutics,
29
30 including improving their translatability and stability, reducing the immunogenicity of mRNAs, and
31
32 efficiently delivering mRNA into targeted cells.1, 3, 20-21 Translatability and stability has been increased
33
34
by optimal modification of mRNAs. Some inherent immunogenicity has been suppressed through
35 removal of double stranded mRNA by HPLC purification.21-22 Like other nucleic acids, the safe and
36
20-21, 23
37 efficient delivery of mRNA is one of the main unmet challenges for mRNA drugs. Single
38
39 stranded mRNAs are less stable than double stranded DNA or siRNA;24-25 thus, there are more stringent
40
41 requirements of suitable carriers to protect mRNA from degradation by endonucleases and facilitate its
42
uptake into target cells. There has been significant progress in both fundamental and translation research
43
44 of DNA and siRNA delivery in the past decade.13, 26-27 However, studies on systemic delivery of mRNA
45
46 are still limited.2-3, 24, 28
47
48
49 A variety of carrier types, most prominently cationic polymers and cationic lipid-based
50 nanoparticles (LNPs), have found utility in gene delivery. LNPs represent an advanced delivery platform
51
52 for siRNA therapeutics and are currently in human clinical trials. LNPs have been recently adapted for
53
54 mRNA delivery in vivo.4, 8, 12, 21, 28-35
Adapting LNPs for longer RNA cargo has required a new
55
56 understanding of LNPs.17, 30-31 Although cationic polymers have been used widely for decades as carriers
57
for long plasmid DNA (pDNA), research as mRNA carriers has only recently been investigated. Given
58
59
60 2
ACS Paragon Plus Environment
Page 3 of 24 Biomacromolecules

1
2
3
that they are already optimized for long nucleic acids, polymers have great potential for mRNA
4
5 delivery.36-37 Positively charged polymers possess controllable structures and diverse functionalities.
6
7 Therefore, cationic polymers have been investigated as another type of candidate for mRNA delivery.25,
8 32, 38-43
9 For example, polyethyleneimine (PEI) or poly(2-dimethylaminoethyl methacrylate)
10
11
(PDMAEMA) based polymers were able to encapsulate mRNA and there is efficient expression of
12 luciferase or GFP in cells after treatment with polymer-mRNA polyplex nanoparticles.38-39 But the
13
14 clinical translation of these polymers may be limited by their poor degradability. A series of
15
16 polyaspartaminde carriers modified with PEI, PEG, or 1,2-diaminoethane was also recently employed
17
18 for the efficient mRNA delivery on a variety of disease models including neurological disorders,
19 osteoarthritis, Alzheimer’s disease, and pancreatic cancer.40-41 mRNA delivery using
20
21 poly(glycoamidoamine) brush polymers was also reported.43 Recent research showed that charge-
22
23 altering releasable transporters are capable of efficacious luciferase mRNA delivery to cultured cells and
24
25 animals.44 Current progress suggested the feasibility of delivering synthetic mRNA for the target protein
26
27
expression in cells. Nevertheless, investigation of mRNA delivery carriers is still limited compared to
28 siRNA and pDNA. Delivery carriers must be further improved according to the unique physiochemical
29
30 properties of single stranded mRNA. In particular, degradability and functionality of polymeric vehicles
31
32 need to be comprehensively considered to meet the requirements in safety, in vivo stabilization, and
33
34 efficacy.
35
36 We recently reported a facile approach for the scalable preparation of functional polyesters with
37
38 tunable molecular weight by the polycondensation of trimethylolpropane allyl ether and diacyl
39
40 chlorides.45-47 Combinatorial modification of the side chain –ene group allows for construction of
41
42 polyesters with diverse chemical functionality and physical properties. Previous studies have suggested
43 that the versatile platform was efficacious for short RNA delivery both in vitro and in vivo.46-47
44
45 Remarkably, high throughput screening of these functional polyesters enabled the discovery of cancer
46
47 selective siRNA nanoparticles that do not require any targeting modification, replying only on
48
49 physicochemical properties of the polymers themselves to mediate cancer selective uptake.46
50
51 In this paper, we aimed to tackle the delivery challenge of emerging mRNA therapeutics using
52
53 the unique combination of functionality and degradability of this polymer platform (Figures 1 and S1).
54
55 We discovered efficient delivery carriers by in vitro screening of the polymer library for mRNA delivery
56
57 to IGROV1 ovarian cancer cells. Candidate carriers were further formulated with Pluronic F-127 to
58
59
60 3
ACS Paragon Plus Environment
Biomacromolecules Page 4 of 24

1
2
3
enhance the stability of mRNA polyplex nanoparticles for in vivo use. Formulated mRNA nanoparticles
4
5 mediated efficacious luciferase expression in the lungs after intravenous administration to mice. Due to
6
7 the facile preparation, tunable molecular weight, degradability and diverse functionality, these functional
8
9 polyesters are suitable carriers for in vitro and in vivo delivery of mRNA therapeutics.
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 Figure 1. (a) A series of degradable functional polyesters was created by combinatorial modification of
36
37 –ene containing polyesters with 20 amino thiols and 3 alkyl thiols. See Figure S1 for all chemical
38 structures. (b) Functional polyesters enable mRNA delivery to cells, mediating efficient expression of
39
40 target proteins in cytoplasm.
41
42
43
44
45
Materials and Methods
46
47
48 Materials. All chemicals for polymer synthesis and modification were purchased from Sigma-Aldrich
49
50 or Fisher Scientific and used as received. Luciferase and mCherry mRNA were purchased from Tri-Link
51
52 Biotechnologies. RPMI-1640, fetal bovine serum (FBS), Dulbecco’s modified phosphate buffered saline
53 (PBS), Trypsin-EDTA (0.25%), and PEO101-PPO56-PEO101 (Pluronic F127, Mw = 12,600, PDI = 1.05)
54
55 were purchased from Sigma-Aldrich. PEG 2000 DMG lipid (Sunbright GM-020, 1,2-dimyristoyl-sn-
56
57 glycerol, methoxypolyethylene glycol) was purchased from NOF America. The Quant-iT RiboGreen
58
59
60 4
ACS Paragon Plus Environment
Page 5 of 24 Biomacromolecules

1
2
3
RNA assay kit and RNAiMax were purchased from Life Technologies and used following the
4
5 manufacturer’s protocols. The ONE-Glo + Tox luciferase assay kit was purchased from Promega. D-
6
7 Luciferin firefly, sodium salt monohydrate was purchased from Gold Biotechnology.
8
9
10 Preparation and characterization of mRNA polyplex nanoparticles. The functional polyesters were
11 synthesized according to our previous papers.45-47 Unmodified polyesters were prepared via the
12
13 polycondensation of trimethylolpropane allyl ether (TPAE) and suberoyl chloride (diACl-C8) with
14
15 pyridine as catalyst in dichloromethane (DCM).45 The polyesters were functionalized through the thiol-
16
17 ene reaction with different amino and alkyl thiols at varying amino/alkyl molar ratios in DMF or DMSO
18
19
using previously reported conditions.46 mRNA polyplex nanoparticles for in vitro studies were prepared
20 by mixing functional polymers (3 g/L in DMSO) with mRNA (citric acid/trisodium citrate buffer, pH
21
22 4.2, 10 mM) in 96-well plates at a polymer/mRNA ratio of 30:1 (wt/wt) and final mRNA concentration
23
24 of 1.25 ng/µL. The polyplex nanoparticles were incubated on a shaking plate (30 rpm) for 30 min prior
25
26 to characterization or addition to cells. To prepare mRNA nanoparticles for in vivo studies, 5 wt% F-127
27 was added to the polymer DMSO solution. Then, the mixture was added into mRNA solution followed
28
29 by homogenization and dialysis (cut-off MW of 3,500 Da) against PBS for 4 h prior to injection to mice.
30
31 The Quant-iT RiboGreen RNA assay kit was used to quantify mRNA binding according to the
32
33 manufacture’s protocols. The size and surface charge of the nanoparticles were measured using a
34
Zetasizer Nano ZS (Malvern, He-Ne, λ = 632 nm). All measurements were performed in quadruplicate
35
36 and the average with standard deviation was reported.
37
38
39 In vitro delivery of mRNA. IGROV1 ovarian cancer cells were cultured in RPMI-1640 medium
40
41 supplemented with 5% FBS (37 oC, 5% CO2). For in vitro screening assays, IGROV1 cells were seeded
42
into opaque white 96-well plates (10,000 cells/well) and incubated for 24 h. The medium was replaced
43
44 with the fresh RPMI-1640 medium with 5% FBS (200 µL/well) and 20 µL luciferase mRNA (Luc
45
46 mRNA) polyplex nanoparticles (25 ng mRNA/well, 181 pM) were added to cells. After incubation for
47
48 24 h, the media was gently removed. The cell viability and luciferase expression were measured using
49
50
ONE-Glo + Tox luciferase assay kits and normalized to untreated cells. All transfection assays were
51 performed in triplicate and the average with standard deviation was reported. RNAiMax positive control
52
53 experiments were conducted following the manufacturer’s protocol. For in vitro fluorescence and
54
55 luminance imaging, IGROV1 cells were seeded into transparent 24-well plates (40,000 cells/well) and
56
57 incubated overnight. Similarly, nanoparticles with 100-800 ng mRNA were added into cells with 800 µL
58
59
60 5
ACS Paragon Plus Environment
Biomacromolecules Page 6 of 24

1
2
3
fresh RPMI-1640 medium (5% FBS). After 24 h transfection, the mCherry or luciferase signal was
4
5 evaluated using an EVOS FL microscope (AMG) or IVIS Lumina imaging system (Caliper Life
6
7 Science). Luc mRNA NPs treated cells were collected and the luciferase expression were quantitated
8
9 using a Luciferase Assay System kit (Promega) according to the manufacture’s protocols.
10
11 In vivo delivery of luciferase mRNA. All animal experiments were approved by the Institutional
12
13 Animal Care and Use Committee (IACUC) of the University of Texas Southwestern Medical Center and
14
15 were consistent with local, state and federal regulations as applicable. NOD SCID mice were purchased
16
17 from the UT Southwestern animal breeding core. Luc mRNA loaded nanoparticles with selected
18
19
functional polyesters and F127 were prepared as described above. 150 µL of mRNA NPs (20 µg Luc
20 mRNA) were administered to NOD SCID mice via tail vein injection (n=4). After 24 h, 80 µL of D-
21
22 luciferin PBS solution (40 g/L) was injected subcutaneously in the neck scruff 10 min prior to
23
24 bioluminescence imaging on an IVIS Lumina imaging system. Ex vivo imaging on systemic organs was
25
26 performed immediately after live animal imaging.
27
28
29
30
31 Results and Discussion
32
33
34
In vitro screening of functional polyesters for mRNA delivery. Nucleic acid delivery involves various
35 steps including the formation of nanoparticles, endocytosis, intracellular trafficking and release, which
36
37 requires a subtle balance among physicochemical properties of carriers, particularly electrostatic binding
38
39 and stabilizing hydrophobicity.3, 17, 46-56
Due to the challenges of identifying efficacious carriers,
40
41 combinatorial high throughput screening has proven to be an efficient way to identify materials for
42 siRNA and plasmid DNA delivery.57-59 Therefore, we constructed a robust library containing 480
43
44 functional polyesters with degradability and diverse chemical functional groups, and used a similar
45
46 screening strategy for the discovery of carriers for mRNA delivery. In vitro studies (Figures 2 and S2)
47
48 show that some mRNA polyplex nanoparticles enable high luciferase expression in IGROV1 cells,
49
indicating efficient luciferase mRNA delivery by these polymers. Particularly, A17 (cysteamine)
50
51 modified polyesters had promising delivery potency. This functional group was also active in our prior
52
53 study of siRNA carriers, suggesting that the same pKa may be suitable for siRNA delivery and mRNA
54
55 delivery. Comparing the overall results to our prior siRNA delivery screen,46 we identified a smaller
56
57
58
59
60 6
ACS Paragon Plus Environment
Page 7 of 24 Biomacromolecules

1
2
3
number of effective polymeric carriers for mRNA delivery, implying that the RNAs with different
4
5 lengths and properties (single versus double stranded) offer different challenges for delivery.
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 Figure 2. In vitro screening of functional polyesters for luciferase mRNA (Luc mRNA) delivery to
38
39 IGROV1 cells enabled identification of efficacious mRNA carriers. Functional polyesters with
40
41 molecular weight of 4,200 g/mol (PE4K) and 8,300 g/mol (PE8K) were functionalized with amino thiols
42
43 (A1-A20) and alkyl thiols (C6-C12) at different molar feed ratios (alkyl thiol : amino thiol) of 1:4, 1:2,
44 1:1, and 2:1. See Figure S1. The functional polyesters were named by the molecular weight of polyester
45
46 followed by the amino thiol, the molar fraction of alkyl thiols, and the alkyl thiol in the modification.
47
48 The Luc mRNA dose was 25 ng mRNA/well (181 pM). The luciferase signal was normalized to
49
50 untreated cells.
51
52
53
54
55
56
57
58
59
60 7
ACS Paragon Plus Environment
Biomacromolecules Page 8 of 24

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 Figure 3. Fluorescence images of IGROV1 cells after 24 h-treatment with mCherry mRNA polyplex
30
31 nanoparticles verified the successful mRNA delivery by selected polymers with varying mRNA dose
32
33 (100, 200, 400, and 800 ng/well). Scale bars = 200 µL.
34
35
36
37
38 To validate the mRNA delivery indicated by the normalized luciferase expression (Figure 2),
39
40 fluorescence imaging of IGROV1 cells was performed after 24 h-treatment with mCherry mRNA NPs.
41
42 Figure 3 shows high mCherry signal under a regular measurement setting (60% intensity), proving
43
successful mCherry mRNA delivery by selected functional polymers. For PE4K-A17-0.33C6 and
44
45 PE4K-A17-0.2C12, the mCherry signal was strong and overall dose responsive (except for a slight
46
47 decrease at 2178 pM for PE4K-A17-0.33C6 due to toxicity or possible aggregation at high
48
49 concentration). There was very limited mCherry expression at low mRNA dose when the polymer chain
50
51 length doubled (PE8K-A17-0.33C6). But mCherry protein expression increased after the mRNA dose
52 was increased to 1272 pM (400 ng/well). It is worth noting that the mass concentration of mRNA in
53
54 screening studies (Figure 2) is identical to the lowest does in 24-well plates. We speculate that
55
56 additional polymer hits could be identified through examination of polymers with different molecular
57
58 weight, or if the mRNA screening dose were to be increased. For the functional polyesters modified with
59
60 8
ACS Paragon Plus Environment
Page 9 of 24 Biomacromolecules

1
2
3
1-(4-(2-hydroxyethyl)piperazin-1-yl)-4-mercaptobutan-1-one (A5), the mCherry expression was
4
5 relatively low for all doses, indicating the predominant role of the side chain amine for cellular uptake
6
7 and endosomal release of mRNA. Overall, this approach allowed identification of efficient mRNA
8
9 delivery carriers via high throughput screening of the versatile polymer library. Focusing on these potent
10
11
polymers, we next evaluated effects of Pluronic F127 formulation on physicochemical properties of
12 mRNA polyplex NPs and the efficacy of mRNA delivery because the formulation with amphiphilic
13
14 stabilizers has been shown to improve the in vivo stability of functional polyester nucleic acid NPs.32, 47
15
16
17 Effects of Pluronic F127 on mRNA delivery. PPO segments of Pluronic F127 could insert into the
18
19
hydrophobic domain and leave hydrophilic PEG on the surface of formulated mRNA polyplex NPs
20 which benefits nucleic acid delivery in vivo by protecting NPs from protein adsorption and aggregation
21
22 at high concentration. On the other hand, too much PEG on the surface may reduce NP-cell interactions
23
24 and cellular uptake.47 To find optimal formulation conditions for mRNA delivery, we evaluated Luc
25
26 mRNA delivery efficacy of NPs formulated with 5 or 10% F127 or PEG2000 DMG lipid (Figures 4 and
27 S3). The addition of F127 did not alter the cell viability, but did lead to a decrease in luciferase
28
29 expression of IGROV1 cell with increased F127 content for some formulations. Without addition of
30
31 F127, some functional polyesters showed comparable mRNA delivery capability to the commercial
32
33 reagent RNAiMax. In contrast to our prior work with F127-stabilized siRNA NPs, where activity was
34
maintained, there was drop in delivery efficacy of the formulated NPs with 10% F127 for most of the
35
36 lead polyesters. Our previous study showed that 5% PEG2000 DMG lipid provided efficient shielding
37
38 for siRNA polyplex nanoparticles without compromising delivery efficacy.47 However, formulation of
39
40 mRNA NPs with a PEG2000 DMG lipid induced a decrease over 2 orders of magnitude (Figure S3),
41
42 which illustrates the challenge of delivering much longer RNAs that are more challenging to load and
43 stabilize. Due to the potential retention or loss of activity,32, 47 the high-throughput screen of polyplexes
44
45 was conducted without stabilizers. Nevertheless, the addition of F127 is essential for in vivo delivery
46
47 because it can improve tolerability and more importantly, greatly improves NP stability in vivo. Among
48
49 the three conditions tested (5% F127, 10% F127, and 5% PEG2000 DMG), 5% F127 was selected for
50
use in studies in vivo because it most optimally balanced delivery efficacy and NP stability. The dose
51
52 response of selected NPs with and without F127 formulation (Figure 4b) further confirmed the mRNA
53
54 delivery to IGROV1 cells.
55
56
57
58
59
60 9
ACS Paragon Plus Environment
Biomacromolecules Page 10 of 24

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 Figure 4. Effects of Pluronic F127 on Luc mRNA delivery to IGROV1 cells. (a) F127 content affects
47
48 the delivery efficacy (50 ng/well, 363 pM). (b) Dose response of Luc mRNA NPs (mean ± s.d., n = 3).
49
50
51
52
53
54
55
56
57
58
59
60 10
ACS Paragon Plus Environment
Page 11 of 24 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
Figure 5. Physicochemical properties of mRNA polyplex nanoparticles with selected polyesters. (a)
51 mRNA binding, (b) size (Dh), and (c) surface charge (ζ) (mean ± s.d., n = 3).
52
53
54 Next, the binding capability of the lead polyesters with mRNA and the resulting properties of
55
56 polyplex NPs were measured. Figure 5a shows that all selected A17 modified polyesters completely
57
58
bound mRNA under the preparation conditions (pH 4.2, 10 mM, and polymer/mRNA weight ratio of
59
60 11
ACS Paragon Plus Environment
Biomacromolecules Page 12 of 24

1
2
3
30:1). The flexibility of single stranded mRNA may benefit the charge neutralization between cationic
4
5 polymers and negatively charged nucleic acids. Most of the selected NPs were smaller than 200 nm. The
6
7 formulation with F127 did not change NP size of C6 and C12 modified polymers, but did decrease the
8
9 size of NPs of C10 polymers (Figure 5b). The diameter of NPs with or without F127 remained constant
10
11
within 0.5 to 2 hours of monitoring prior to addition to cells. Furthermore, the surface charge of mRNA
12 NPs with selected polyesters dramatically decreased after addition of F127, indicating the formation of
13
14 the protective PEG layer on the NP surface (Figure 5c). Meanwhile, the formulation-induced charge
15
16 decrease weakened NP interactions with cell surfaces, resulting in decrease in mRNA delivery in vitro
17
18 for some formulations (Figure 4a).
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
Figure 6. Luminescence images of IGROV1 cells after 24 h-treatment with Luc mRNA polyplex
52 nanoparticles at varying mRNA dose (100, 200, 400, and 800 ng/well).
53
54
55
56
57
58
59
60 12
ACS Paragon Plus Environment
Page 13 of 24 Biomacromolecules

1
2
3
Luminescence imaging of cells treated with Luc mRNA NPs. Luciferase encoded mRNA is powerful
4
5 for mRNA delivery evaluation because of the convenient luminescence measurement both in vitro and
6
7 in vivo. Prior to the administration of mRNA NPs to mice, in vitro luminescence imaging was performed
8
9 on IGROV1 cells for the selected formulations (Figure 6). For cases of NPs without F127, the
10
11
luminescence signal decreased at highest mRNA dose due to the cytotoxicity and possible aggregation.
12 After formulation with 5% F127, the luminescence of cells was lower than that without formulation at
13
14 low mRNA dose. But the luciferase expression was stronger at high dose due to the improvement in NP
15
16 stability. Further increase of F127 content to 10% led to a decrease in luminescence, suggesting
17
18 inefficient Luc mRNA delivery by polyesters with 10% F127.
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
Figure 7. In vivo efficacy of mRNA NPs delivered by tail vein injection to mice at a dose of 1 mg/kg
55
56 mRNA. (a) Lead Luc mRNA NPs enable efficient mRNA delivery to mice. (b) Representative ex vivo
57
58
59
60 13
ACS Paragon Plus Environment
Biomacromolecules Page 14 of 24

1
2
3
images show luciferase expression mainly in the lungs (minor spleen expression) after systemic
4
5 administration of Luc mRNA NPs with PE4K-A17-0.33C12-5% F127.
6
7
8
9
10
11
In vivo evaluation of mRNA delivery. Based on the promising in vitro luminescence imaging (Figure
12 6), we focused on 5% F127 formulated NPs in studies in vivo. There was clear luciferase expression in
13
14 mice after i.v. administration of Luc mRNA NPs with 5% F127. When F127 content increased to 10%,
15
16 there was almost no luciferase detected under the same measurement setting (Figure 7a), in agreement
17
18 with reduced in vitro delivery efficacy observed with higher F127 content. Luminescence images of
19 excised organs following i.v. injection of Luc mRNA NPs with PE4K-A17-0.33C12 (Figure S4) and 5%
20
21 F127 demonstrate luciferase expression predominantly in the lungs, with less delivery to the spleen
22
23 (Figure 7b). The mixture of mRNA and functional polyesters without F127 became turbid immediately
24
25 under the in vivo formulation concentration (0.13 g mRNA/L) and there was a precipitate on the bottom
26
of the dialysis cup after 24 hour dialysis against PBS. Therefore, un-formulated NPs (i.e. NPs without
27
28 the F127 stabilizer) were not included in the in vivo studies. A large number of studies showed that
29
30 nucleic acid NPs readily accumulate in the liver after i.v. administration due to the size of nanoparticles
31
32 and possible adsorption of hepatocyte targeting proteins in serum (e.g. ApoE).60 It has been shown that
33
34
altering administration method to aerosol inhalation enables specific delivery of siRNA or mRNA in the
35 lungs.24, 46 Our results indicate that functional polyesters enable efficient mRNA delivery to the lungs
36
37 but not the liver via i.v. injection, which is consistent with data on poly(β-amino ester) (PBAE)
38
39 polymers in a recent report.32 It was shown that PEGylated mRNA polyplex NPs accumulated in
40
41 multiple organs (including liver) but that the mRNA was only translated to protein in the lungs.32 It was
42
further speculated that specific protein adsorption or physiology may play a role in lung delivery.32, 60-63
43
44 Moreover, a report on a degradable LNP indicated preferential delivery to the spleen, hypothesized to be
45
46 due to the degradable bonds.64 Thus, factors including surface charge, size, and degradability might
47
48 contribute to lung mRNA delivery of degradable mRNA NPs. As shown in luminescence images,
49
50
formulation with additional F127 (10%) diminished luciferase activity. Ex vivo organ images from mice
51 injected with PE4K-A17-0.33C12 - 5% F127 NPs show more than 30-fold higher luciferase expression
52
53 in the lungs that mRNA NPs with PE4K-A17-0.2C12 -10% F127 (Figure S5). Inclusion of additional
54
55 F127 may provide better protection from the aggregation or precipitation, but it also sacrifices the
56
57
58
59
60 14
ACS Paragon Plus Environment
Page 15 of 24 Biomacromolecules

1
2
3
mRNA delivery potency. The balance between these two factors should be carefully considered in the
4
5 design and formulation of nucleic acid NPs.
6
7
8 Conclusions
9
10
11
In this paper, we report a robust functional polyester library for the discovery of efficient delivery
12 carriers for emerging mRNA therapeutics. In vitro screening of the polyester library enabled
13
14 identification of amine A17 modified polyester series as being potent carriers for Luc mRNA delivery to
15
16 IGROV1 cells. Successful delivery was validated by fluorescence imaging of cells treated with polyplex
17
18 NPs of mCherry mRNA and selected polyesters. To improve the stability of mRNA NPs at high
19 concentration and in serum, formulated mRNA NPs were prepared by adding 5 or 10% triblock
20
21 copolymer PEO101-PPO56-PEO101 (F127) into the stock solution of the lead functional polyesters prior to
22
23 mixing with mRNA. Formulation with F127 led to a slight decrease in the size and a remarkable
24
25 decrease in surface charge of mRNA NPs depending on the polyester and F127 content. It did not
26
sacrifice the encapsulation of mRNA into polyplex NPs, but the delivery efficacy decreased with higher
27
28 F127 content. Luminescence imaging showed that the formulation with 5% F127 enabled efficient
29
30 delivery of Luc mRNA at high dose whereas addition of 10% F127 significantly decreased the potency
31
32 across the entire dose range. To balance the protective effect and delivery potency, formulated mRNA
33
34
NPs with moderate F127 coating (5%) were examined in vivo. Bioluminescence imaging demonstrated
35 efficient Luc mRNA delivery to mice after intravenous injection of NPs, where the luciferase expression
36
37 was mainly in the lungs. The safe and efficient mRNA delivery specifically to lungs by functional
38
39 polyesters may hold potential for the treatment of pulmonary diseases.
40
41
42
Acknowledgements
43
44 D.J.S. gratefully acknowledges financial support from the Cancer Prevention and Research Institute of
45
46 Texas (CPRIT) (R1212), the Welch Foundation (I-1855), the American Cancer Society (RSG-17-012-
47
48 01), and the Mary Kay Foundation (049-15). Y.Y. gratefully acknowledges financial support from
49
Zhejiang University of Technology (105008529).
50
51
52 Supporting Information
53
54
55 The Supporting Information is available free of charge on the ACS Publications website at DOI:
56
57
10.1021/acs.biomac.
58
59
60 15
ACS Paragon Plus Environment
Biomacromolecules Page 16 of 24

1
2
3
This information includes the chemical structures of functional polyesters modified with amino- and
4
5 alkyl-thiols used in the study, cell viability after treatment with mRNA polyplex NPs, effects of
6
7 PEG2000 DMG lipid on mRNA delivery, and ex vivo quantification of luminescence intensity in organs
8
9 after administration of mRNA polyplex NPs
10
11
References
12
13
14 1. Sahin, U.; Kariko, K.; Tureci, O., mRNA-based therapeutics: Developing a new class of drugs.
15
16 Nat. Rev. Drug Discovery 2014, 13 (10), 759-780.
17
18 2. Kauffman, K. J.; Webber, M. J.; Anderson, D. G., Materials for non-viral intracellular delivery
19 of messenger RNA therapeutics. J. Controlled Release 2016, 240, 227-234.
20
21 3. Hajj, K. A.; Whitehead, K. A., Tools for translation: non-viral materials for therapeutic mRNA
22
23 delivery. Nat. Rev. Mater. 2017, 2 (10), 17056.
24
25 4. Kormann, M. S.; Hasenpusch, G.; Aneja, M. K.; Nica, G.; Flemmer, A. W.; Herber-Jonat, S.;
26
Huppmann, M.; Mays, L. E.; Illenyi, M.; Schams, A.; Griese, M.; Bittmann, I.; Handgretinger, R.; Hartl,
27
28 D.; Rosenecker, J.; Rudolph, C., Expression of therapeutic proteins after delivery of chemically
29
30 modified mRNA in mice. Nat. Biotechnol. 2011, 29 (2), 154-157.
31
32 5. Petsch, B.; Schnee, M.; Vogel, A. B.; Lange, E.; Hoffmann, B.; Voss, D.; Schlake, T.; Thess, A.;
33
34
Kallen, K. J.; Stitz, L.; Kramps, T., Protective efficacy of in vitro synthesized, specific mRNA vaccines
35 against influenza A virus infection. Nat. Biotechnol. 2012, 30 (12), 1210-1216.
36
37 6. Zangi, L.; Lui, K. O.; von Gise, A.; Ma, Q.; Ebina, W.; Ptaszek, L. M.; Spater, D.; Xu, H.;
38
39 Tabebordbar, M.; Gorbatov, R.; Sena, B.; Nahrendorf, M.; Briscoe, D. M.; Li, R. A.; Wagers, A. J.;
40
41 Rossi, D. J.; Pu, W. T.; Chien, K. R., Modified mRNA directs the fate of heart progenitor cells and
42
induces vascular regeneration after myocardial infarction. Nat. Biotechnol. 2013, 31 (10), 898-907.
43
44 7. Miller, J. D.; Ganat, Y. M.; Kishinevsky, S.; Bowman, R. L.; Liu, B.; Tu, E. Y.; Mandal, P. K.;
45
46 Vera, E.; Shim, J. W.; Kriks, S.; Taldone, T.; Fusaki, N.; Tomishima, M. J.; Krainc, D.; Milner, T. A.;
47
48 Rossi, D. J.; Studer, L., Human iPSC-based modeling of late-onset disease via progerin-induced aging.
49
50
Cell Stem Cell 2013, 13 (6), 691-705.
51 8. Thess, A.; Grund, S.; Mui, B. L.; Hope, M. J.; Baumhof, P.; Fotin-Mleczek, M.; Schlake, T.,
52
53 Sequence-engineered mRNA Without Chemical Nucleoside Modifications Enables an Effective Protein
54
55 Therapy in Large Animals. Mol. Ther. 2015, 23 (9), 1456-1464.
56
57
58
59
60 16
ACS Paragon Plus Environment
Page 17 of 24 Biomacromolecules

1
2
3
9. Chahal, J. S.; Khan, O. F.; Cooper, C. L.; McPartlan, J. S.; Tsosie, J. K.; Tilley, L. D.; Sidik, S.
4
5 M.; Lourido, S.; Langer, R.; Bavari, S.; Ploegh, H. L.; Anderson, D. G., Dendrimer-RNA nanoparticles
6
7 generate protective immunity against lethal Ebola, H1N1 influenza, and Toxoplasma gondii challenges
8
9 with a single dose. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (35), E4133–E4142.
10
11
10. Kranz, L. M.; Diken, M.; Haas, H.; Kreiter, S.; Loquai, C.; Reuter, K. C.; Meng, M.; Fritz, D.;
12 Vascotto, F.; Hefesha, H.; Grunwitz, C.; Vormehr, M.; Husemann, Y.; Selmi, A.; Kuhn, A. N.; Buck, J.;
13
14 Derhovanessian, E.; Rae, R.; Attig, S.; Diekmann, J.; Jabulowsky, R. A.; Heesch, S.; Hassel, J.;
15
16 Langguth, P.; Grabbe, S.; Huber, C.; Tureci, O.; Sahin, U., Systemic RNA delivery to dendritic cells
17
18 exploits antiviral defence for cancer immunotherapy. Nature 2016, 534 (7607), 396-401.
19 11. Alberer, M.; Gnad-Vogt, U.; Hong, H. S.; Mehr, K. T.; Backert, L.; Finak, G.; Gottardo, R.; Bica,
20
21 M. A.; Garofano, A.; Koch, S. D.; Fotin-Mleczek, M.; Hoerr, I.; Clemens, R.; von Sonnenburg, F.,
22
23 Safety and immunogenicity of a mRNA rabies vaccine in healthy adults: an open-label, non-randomised,
24
25 prospective, first-in-human phase 1 clinical trial. Lancet 2017, 390 (10101), 1511-1520.
26
27
12. Oberli, M. A.; Reichmuth, A. M.; Dorkin, J. R.; Mitchell, M. J.; Fenton, O. S.; Jaklenec, A.;
28 Anderson, D. G.; Langer, R.; Blankschtein, D., Lipid nanoparticle assisted mRNA delivery for potent
29
30 cancer immunotherapy. Nano Lett. 2017, 17 (3), 1326-1335.
31
32 13. Yin, H.; Kanasty, R. L.; Eltoukhy, A. A.; Vegas, A. J.; Dorkin, J. R.; Anderson, D. G., Non-viral
33
34 vectors for gene-based therapy. Nat. Rev. Genet. 2014, 15 (8), 541-555.
35 14. Hendel, A.; Bak, R. O.; Clark, J. T.; Kennedy, A. B.; Ryan, D. E.; Roy, S.; Steinfeld, I.; Lunstad,
36
37 B. D.; Kaiser, R. J.; Wilkens, A. B.; Bacchetta, R.; Tsalenko, A.; Dellinger, D.; Bruhn, L.; Porteus, M.
38
39 H., Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells.
40
41 Nat. Biotechnol. 2015, 33 (9), 985-989.
42
43
15. Wang, J. B.; Exline, C. M.; DeClercq, J. J.; Llewellyn, G. N.; Hayward, S. B.; Li, P. W. L.;
44 Shivak, D. A.; Surosky, R. T.; Gregory, P. D.; Holmes, M. C.; Cannon, P. M., Homology-driven
45
46 genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat.
47
48 Biotechnol. 2015, 33 (12), 1256–1263.
49
50 16. Wang, J.; DeClercq, J. J.; Hayward, S. B.; Li, P. W.; Shivak, D. A.; Gregory, P. D.; Lee, G.;
51 Holmes, M. C., Highly efficient homology-driven genome editing in human T cells by combining zinc-
52
53 finger nuclease mRNA and AAV6 donor delivery. Nucleic Acids Res. 2016, 44 (3), e30.
54
55
56
57
58
59
60 17
ACS Paragon Plus Environment
Biomacromolecules Page 18 of 24

1
2
3
17. Miller, J. B.; Zhang, S.; Kos, P.; Xiong, H.; Zhou, K.; Perelman, S. S.; Zhu, H.; Siegwart, D. J.,
4
5 Non-viral CRISPR/Cas gene editing in vitro and in vivo enabled by synthetic nanoparticle co-delivery of
6
7 Cas9 mRNA and sgRNA. Angew. Chem. Int. Ed. 2017, 56 (4), 1059-1063.
8
9 18. Jiang, C.; Mei, M.; Li, B.; Zhu, X. R.; Zu, W. H.; Tian, Y. J.; Wang, Q. N.; Guo, Y.; Dong, Y. Z.;
10
11
Tan, X., A non-viral CRISPR/Cas9 delivery system for therapeutically targeting HBV DNA and pcsk9
12 in vivo. Cell Res 2017, 27 (3), 440-443.
13
14 19. Hacein-Bey-Abina, S.; von Kalle, C.; Schmidt, M.; Le Deist, F.; Wulffraat, N.; McIntyre, E.;
15
16 Radford, I.; Villeval, J. L.; Fraser, C. C.; Cavazzana-Calvo, M.; Fischer, A., A serious adverse event
17
18 after successful gene therapy for X-linked severe combined immunodeficiency. New Engl. J. Med. 2003,
19 348 (3), 255-256.
20
21 20. Weissman, D., mRNA transcript therapy. Expert Review of Vaccines 2015, 14 (2), 265-281.
22
23 21. Pardi, N.; Tuyishime, S.; Muramatsu, H.; Kariko, K.; Mui, B. L.; Tam, Y. K.; Madden, T. D.;
24
25 Hope, M. J.; Weissman, D., Expression kinetics of nucleoside-modified mRNA delivered in lipid
26
27
nanoparticles to mice by various routes. J. Controlled Release 2015, 217, 345-351.
28 22. Kariko, K.; Muramatsu, H.; Ludwig, J.; Weissman, D., Generating the optimal mRNA for
29
30 therapy: HPLC purification eliminates immune activation and improves translation of nucleoside-
31
32 modified, protein-encoding mRNA. Nucleic Acids Res. 2011, 39 (21), e142.
33
34 23. Guan, S.; Rosenecker, J., Nanotechnologies in delivery of mRNA therapeutics using nonviral
35 vector-based delivery systems. Gene Ther. 2017, 24 (3), 133-143.
36
37 24. Jarzebinska, A.; Pasewald, T.; Lambrecht, J.; Mykhaylyk, O.; Kummerling, L.; Beck, P.;
38
39 Hasenpusch, G.; Rudolph, C.; Plank, C.; Dohmen, C., A single methylene group in oligoalkylamine-
40
41 based cationic polymers and lipids promotes enhanced mRNA delivery. Angew. Chem. Int. Ed. 2016, 55
42
43
(33), 9591-9595.
44 25. Badieyan, Z. S.; Berezhanskyy, T.; Utzinger, M.; Aneja, M. K.; Emrich, D.; Erben, R.; Schuler,
45
46 C.; Altpeter, P.; Ferizi, M.; Hasenpusch, G.; Rudolph, C.; Plank, C., Transcript-activated collagen
47
48 matrix as sustained mRNA delivery system for bone regeneration. J. Controlled Release 2016, 239, 137-
49
50 148.
51 26. Kanasty, R.; Dorkin, J. R.; Vegas, A.; Anderson, D., Delivery materials for siRNA therapeutics.
52
53 Nat. Mater. 2013, 12 (11), 967-977.
54
55
56
57
58
59
60 18
ACS Paragon Plus Environment
Page 19 of 24 Biomacromolecules

1
2
3
27. Davis, M. E.; Zuckerman, J. E.; Choi, C. H.; Seligson, D.; Tolcher, A.; Alabi, C. A.; Yen, Y.;
4
5 Heidel, J. D.; Ribas, A., Evidence of RNAi in humans from systemically administered siRNA via
6
7 targeted nanoparticles. Nature 2010, 464 (7291), 1067-1070.
8
9 28. Fenton, O. S.; Kauffman, K. J.; McClellan, R. L.; Appel, E. A.; Dorkin, J. R.; Tibbitt, M. W.;
10
11
Heartlein, M. W.; DeRosa, F.; Langer, R.; Anderson, D. G., Bioinspired alkenyl amino alcohol ionizable
12 lipid materials for highly potent in vivo mRNA delivery. Adv. Mater. 2016, 28 (15), 2939-2943.
13
14 29. Kariko, K.; Muramatsu, H.; Keller, J. M.; Weissman, D., Increased erythropoiesis in mice
15
16 injected with submicrogram quantities of pseudouridine-containing mRNA encoding erythropoietin. Mol.
17
18 Ther. - Nucl. Acids 2012, 20 (5), 948-953.
19 30. Kauffman, K. J.; Dorkin, J. R.; Yang, J. H.; Heartlein, M. W.; DeRosa, F.; Mir, F. F.; Fenton, O.
20
21 S.; Anderson, D. G., Optimization of lipid nanoparticle formulations for mRNA delivery in vivo with
22
23 fractional factorial and definitive screening designs. Nano Lett. 2015, 15 (11), 7300-7306.
24
25 31. Li, B.; Luo, X.; Deng, B.; Wang, J.; McComb, D. W.; Shi, Y.; Gaensler, K. M.; Tan, X.; Dunn,
26
27
A. L.; Kerlin, B. A.; Dong, Y., An orthogonal array optimization of lipid-like nanoparticles for mRNA
28 delivery in vivo. Nano Lett. 2015, 15 (12), 8099-8107.
29
30 32. Kaczmarek, J. C.; Patel, A. K.; Kauffman, K. J.; Fenton, O. S.; Webber, M. J.; Heartlein, M. W.;
31
32 DeRosa, F.; Anderson, D. G., Polymer-lipid nanoparticles for systemic delivery of mRNA to the lungs.
33
34 Angew. Chem. Int. Ed. 2016, 55 (44), 13808-13812.
35 33. Turnbull, I. C.; Eltoukhy, A. A.; Fish, K. M.; Nonnenmacher, M.; Ishikawa, K.; Chen, J.; Hajjar,
36
37 R. J.; Anderson, D. G.; Costa, K. D., Myocardial delivery of lipidoid nanoparticle carrying modRNA
38
39 induces rapid and transient expression. Mol. Ther. - Nucl. Acids 2016, 24 (1), 66-75.
40
41 34. Li, B.; Luo, X.; Deng, B.; Giancola, J. B.; McComb, D. W.; Schmittgen, T. D.; Dong, Y., Effects
42
43
of local structural transformation of lipid-like compounds on delivery of messenger RNA. Sci. Rep.
44 2016, 6, 22137.
45
46 35. Phua, K. K. L.; Liu, Y.; Sim, S. H., Non-linear enhancement of mRNA delivery efficiencies by
47
48 influenza A derived NS1 protein engendering host gene inhibition property. Biomaterials 2017, 133, 29-
49
50 36.
51 36. Li, J.; Wang, W.; He, Y.; Li, Y.; Yan, E. Z.; Zhang, K.; Irvine, D. J.; Hammond, P. T.,
52
53 Structurally programmed assembly of translation initiation nanoplex for superior mRNA delivery. ACS
54
55 Nano 2017, 11 (3), 2531-2544.
56
57
58
59
60 19
ACS Paragon Plus Environment
Biomacromolecules Page 20 of 24

1
2
3
37. Chen, Q.; Qi, R.; Chen, X.; Yang, X.; Wu, S.; Xiao, H.; Dong, W., A targeted and stable
4
5 polymeric nanoformulation enhances systemic delivery of mRNA to tumors. Mol. Ther. 2017, 25 (1),
6
7 92-101.
8
9 38. Rejman, J.; Tavernier, G.; Bavarsad, N.; Demeester, J.; De Smedt, S. C., mRNA transfection of
10
11
cervical carcinoma and mesenchymal stem cells mediated by cationic carriers. J. Controlled Release
12 2010, 147 (3), 385-391.
13
14 39. Cheng, C.; Convertine, A. J.; Stayton, P. S.; Bryers, J. D., Multifunctional triblock copolymers
15
16 for intracellular messenger RNA delivery. Biomaterials 2012, 33 (28), 6868-6876.
17
18 40. Uchida, H.; Itaka, K.; Nomoto, T.; Ishii, T.; Suma, T.; Ikegami, M.; Miyata, K.; Oba, M.;
19 Nishiyama, N.; Kataoka, K., Modulated protonation of side chain aminoethylene repeats in N-
20
21 substituted polyaspartamides promotes mRNA transfection. J. Am. Chem. Soc. 2014, 136 (35), 12396-
22
23 12405.
24
25 41. Aini, H.; Itaka, K.; Fujisawa, A.; Uchida, H.; Uchida, S.; Fukushima, S.; Kataoka, K.; Saito, T.;
26
27
Chung, U. I.; Ohba, S., Messenger RNA delivery of a cartilage-anabolic transcription factor as a
28 disease-modifying strategy for osteoarthritis treatment. Sci. Rep. 2016, 6, 18743.
29
30 42. Mahiny, A. J.; Dewerth, A.; Mays, L. E.; Alkhaled, M.; Mothes, B.; Malaeksefat, E.; Loretz, B.;
31
32 Rottenberger, J.; Brosch, D. M.; Reautschnig, P.; Surapolchai, P.; Zeyer, F.; Schams, A.; Carevic, M.;
33
34 Bakele, M.; Griese, M.; Schwab, M.; Nurnberg, B.; Beer-Hammer, S.; Handgretinger, R.; Hartl, D.;
35 Lehr, C. M.; Kormann, M. S., In vivo genome editing using nuclease-encoding mRNA corrects SP-B
36
37 deficiency. Nat. Biotechnol. 2015, 33 (6), 584-586.
38
39 43. Dong, Y.; Dorkin, J. R.; Wang, W.; Chang, P. H.; Webber, M. J.; Tang, B. C.; Yang, J.; Abutbul-
40
41 Ionita, I.; Danino, D.; DeRosa, F.; Heartlein, M.; Langer, R.; Anderson, D. G., Poly(glycoamidoamine)
42
43
brushes formulated nanomaterials for systemic siRNA and mRNA delivery in vivo. Nano Lett. 2016, 16
44 (2), 842-848.
45
46 44. McKinlay, C. J.; Vargas, J. R.; Blake, T. R.; Hardy, J. W.; Kanada, M.; Contag, C. H.; Wender,
47
48 P. A.; Waymouth, R. M., Charge-altering releasable transporters (CARTs) for the delivery and release of
49
50 mRNA in living animals. Proc. Natl. Acad. Sci. U.S.A. 2017, 114 (4), E448-E456.
51 45. Yan, Y. F.; Siegwart, D. J., Scalable synthesis and derivation of functional polyesters bearing
52
53 ene and epoxide side chains. Polym. Chem. 2014, 5 (4), 1362-1371.
54
55 46. Yan, Y.; Liu, L.; Xiong, H.; Miller, J. B.; Zhou, K.; Kos, P.; Huffman, K. E.; Elkassih, S.;
56
57 Norman, J. W.; Carstens, R.; Kim, J.; Minna, J. D.; Siegwart, D. J., Functional polyesters enable
58
59
60 20
ACS Paragon Plus Environment
Page 21 of 24 Biomacromolecules

1
2
3
selective siRNA delivery to lung cancer over matched normal cells. Proc. Natl. Acad. Sci. U.S.A. 2016,
4
5 113 (39), E5702-E5710.
6
7 47. Yan, Y.; Zhou, K.; Xiong, H.; Miller, J. B.; Motea, E. A.; Boothman, D. A.; Liu, L.; Siegwart, D.
8
9 J., Aerosol delivery of stabilized polyester-siRNA nanoparticles to silence gene expression in orthotopic
10
11
lung tumors. Biomaterials 2017, 118, 84-93.
12 48. Wong, S. Y.; Sood, N.; Putnam, D., Combinatorial evaluation of cations, pH-sensitive and
13
14 hydrophobic moieties for polymeric vector design. Mol. Ther. - Nucl. Acids 2009, 17 (3), 480-490.
15
16 49. Zhi, D.; Zhang, S.; Wang, B.; Zhao, Y.; Yang, B.; Yu, S., Transfection efficiency of cationic
17
18 lipids with different hydrophobic domains in gene delivery. Bioconjugate Chem. 2010, 21 (4), 563-577.
19 50. Liu, Z. H.; Zhang, Z. Y.; Zhou, C. R.; Jiao, Y. P., Hydrophobic modifications of cationic
20
21 polymers for gene delivery. Prog. Polym. Sci. 2010, 35 (9), 1144-1162.
22
23 51. Siegwart, D. J.; Whitehead, K. A.; Nuhn, L.; Sahay, G.; Cheng, H.; Jiang, S.; Ma, M.; Lytton-
24
25 Jean, A.; Vegas, A.; Fenton, P.; Levins, C. G.; Love, K. T.; Lee, H.; Cortez, C.; Collins, S. P.; Li, Y. F.;
26
27
Jang, J.; Querbes, W.; Zurenko, C.; Novobrantseva, T.; Langer, R.; Anderson, D. G., Combinatorial
28 synthesis of chemically diverse core-shell nanoparticles for intracellular delivery. Proc. Natl. Acad. Sci.
29
30 U.S.A. 2011, 108 (32), 12996-3001.
31
32 52. Nelson, C. E.; Kintzing, J. R.; Hanna, A.; Shannon, J. M.; Gupta, M. K.; Duvall, C. L.,
33
34 Balancing cationic and hydrophobic content of PEGylated siRNA polyplexes enhances endosome
35 escape, stability, blood circulation time, and bioactivity in vivo. ACS Nano 2013, 7 (10), 8870-8880.
36
37 53. Hao, J.; Kos, P.; Zhou, K.; Miller, J. B.; Xue, L.; Yan, Y.; Xiong, H.; Elkassih, S.; Siegwart, D.
38
39 J., Rapid synthesis of a lipocationic polyester library via ring-opening polymerization of functional
40
41 valerolactones for efficacious siRNA delivery. J. Am. Chem. Soc. 2015, 137 (29), 9206-9209.
42
43
54. Hao, J.; Elkassih, S.; Siegwart, D. J., Progress towards the synthesis of amino polyesters via
44 ring-opening polymerization (ROP) of functional lactones. Synlett 2016, 27 (16), 2285-2292.
45
46 55. Zhou, K.; Kos, P.; Yan, Y.; Xiong, H.; Min, Y. L.; Kinghorn, K. A.; Minnig, J. T.; Miller, J. B.;
47
48 Siegwart, D. J., Intercalation-mediated nucleic acid nanoparticles for siRNA delivery. Chem. Commun.
49
50 2016, 52 (82), 12155-12158.
51 56. Zhou, K.; Nguyen, L. H.; Miller, J. B.; Yan, Y.; Kos, P.; Xiong, H.; Li, L.; Hao, J.; Minnig, J. T.;
52
53 Zhu, H.; Siegwart, D. J., Modular degradable dendrimers enable small RNAs to extend survival in an
54
55 aggressive liver cancer model. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (3), 520-525.
56
57
58
59
60 21
ACS Paragon Plus Environment
Biomacromolecules Page 22 of 24

1
2
3
57. Lynn, D. M.; Langer, R., Degradable poly(beta-amino esters): Synthesis, characterization, and
4
5 self-assembly with plasmid DNA. J. Am. Chem. Soc. 2000, 122 (44), 10761-10768.
6
7 58. Akinc, A.; Zumbuehl, A.; Goldberg, M.; Leshchiner, E. S.; Busini, V.; Hossain, N.; Bacallado, S.
8
9 A.; Nguyen, D. N.; Fuller, J.; Alvarez, R.; Borodovsky, A.; Borland, T.; Constien, R.; de Fougerolles, A.;
10
11
Dorkin, J. R.; Narayanannair Jayaprakash, K.; Jayaraman, M.; John, M.; Koteliansky, V.; Manoharan,
12 M.; Nechev, L.; Qin, J.; Racie, T.; Raitcheva, D.; Rajeev, K. G.; Sah, D. W.; Soutschek, J.; Toudjarska,
13
14 I.; Vornlocher, H. P.; Zimmermann, T. S.; Langer, R.; Anderson, D. G., A combinatorial library of lipid-
15
16 like materials for delivery of RNAi therapeutics. Nat. Biotechnol. 2008, 26 (5), 561-569.
17
18 59. Yan, Y.; Xue, L.; Miller, J. B.; Zhou, K.; Kos, P.; Elkassih, S.; Liu, L.; Nagai, A.; Xiong, H.;
19 Siegwart, D. J., One-pot synthesis of functional poly(amino ester sulfide)s and utility in delivering
20
21 pDNA and siRNA. Polymer 2015, 72, 271-280.
22
23 60. Akinc, A.; Querbes, W.; De, S.; Qin, J.; Frank-Kamenetsky, M.; Jayaprakash, K. N.; Jayaraman,
24
25 M.; Rajeev, K. G.; Cantley, W. L.; Dorkin, J. R.; Butler, J. S.; Qin, L.; Racie, T.; Sprague, A.; Fava, E.;
26
27
Zeigerer, A.; Hope, M. J.; Zerial, M.; Sah, D. W.; Fitzgerald, K.; Tracy, M. A.; Manoharan, M.;
28 Koteliansky, V.; Fougerolles, A.; Maier, M. A., Targeted delivery of RNAi therapeutics with
29
30 endogenous and exogenous ligand-based mechanisms. Mol. Ther. 2010, 18 (7), 1357-64.
31
32 61. Dahlman, J. E.; Barnes, C.; Khan, O. F.; Thiriot, A.; Jhunjunwala, S.; Shaw, T. E.; Xing, Y. P.;
33
34 Sager, H. B.; Sahay, G.; Speciner, L.; Bader, A.; Bogorad, R. L.; Yin, H.; Racie, T.; Dong, Y. Z.; Jiang,
35 S.; Seedorf, D.; Dave, A.; Sandhu, K. S.; Webber, M. J.; Novobrantseva, T.; Ruda, V. M.; Lytton-Jean,
36
37 A. K. R.; Levins, C. G.; Kalish, B.; Mudge, D. K.; Perez, M.; Abezgauz, L.; Dutta, P.; Smith, L.;
38
39 Charisse, K.; Kieran, M. W.; Fitzgerald, K.; Nahrendorf, M.; Danino, D.; Tuder, R. M.; von Andrian, U.
40
41 H.; Akinc, A.; Panigrahy, D.; Schroeder, A.; Koteliansky, V.; Langer, R.; Anderson, D. G., In vivo
42
43
endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat. Nanotechnol.
44 2014, 9 (8), 648-655.
45
46 62. Khan, O. F.; Zaia, E. W.; Jhunjhunwala, S.; Xue, W.; Cai, W. X.; Yun, D. S.; Barnes, C. M.;
47
48 Dahlman, J. E.; Dong, Y. Z.; Pelet, J. M.; Webber, M. J.; Tsosie, J. K.; Jacks, T. E.; Langer, R.;
49
50 Anderson, D. G., Dendrimer-inspired nanomaterials for the in vivo delivery of siRNA to lung
51 vasculature. Nano Lett. 2015, 15 (5), 3008-3016.
52
53 63. Xue, W.; Dahlman, J. E.; Tammela, T.; Khan, O. F.; Sood, S.; Dave, A.; Cai, W.; Chirino, L. M.;
54
55 Yang, G. R.; Bronson, R.; Crowley, D. G.; Sahay, G.; Schroeder, A.; Langer, R.; Anderson, D. G.; Jacks,
56
57
58
59
60 22
ACS Paragon Plus Environment
Page 23 of 24 Biomacromolecules

1
2
3
T., Small RNA combination therapy for lung cancer. Proc. Natl. Acad. Sci. U.S.A. 2014, 111 (34),
4
5 E3553-E3561.
6
7 64. Fenton, O. S.; Kauffman, K. J.; Kaczmarek, J. C.; McClellan, R. L.; Jhunjhunwala, S.; Tibbitt, M.
8
9 W.; Zeng, M. D.; Appel, E. A.; Dorkin, J. R.; Mir, F. F.; Yang, J. H.; Oberli, M. A.; Heartlein, M. W.;
10
11
DeRosa, F.; Langer, R.; Anderson, D. G., Synthesis and biological evaluation of ionizable lipid
12 materials for the in vivo delivery of messenger RNA to B lymphocytes. Adv. Mater. 2017, 29 (33),
13
14 1606944.
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 23
ACS Paragon Plus Environment
Biomacromolecules Page 24 of 24

1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
Table of Contents (TOC) Image
22
23 465x196mm (300 x 300 DPI)
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 Paragon Plus Environment

You might also like