ArticlePDF Available

Therapeutic Protein PEPylation: The Helix of Nonfouling Synthetic Polypeptides Minimizes Antidrug Antibody Generation

Authors:

Abstract and Figures

Polymer conjugation is a clinically proven approach to generate long acting protein drugs with decreased immune responses. Although poly(ethylene glycol) (PEG) is one of the most commonly used conjugation partners due to its unstructured conformation, its therapeutic application is limited by its poor biodegradability, propensity to induce an anti-PEG immune response, and the resultant accelerated blood clearance (ABC) effect. Moreover, the prevailing preference of unstructured polymers for protein conjugation still lacks strong animal data support with appropriate control reagents. By using two biodegradable synthetic polypeptides with similar structural compositions (l-P(EG3Glu) and dl-P(EG3Glu)) for site-specific protein modification, in the current study, we systematically investigate the effect of the polymer conformation on the in vivo pharmacological performances of the resulting conjugates. Our results reveal that the conjugate l20K-IFN, interferon (IFN) modified with the helical polypeptide l-P(EG3Glu) shows improved binding affinity, in vitro antiproliferative activity, and in vivo efficacy compared to those modified with the unstructured polypeptide analogue dl-P(EG3Glu) or PEG. Moreover, l20K-IFN triggered significantly less antidrug and antipolymer antibodies than the other two. Importantly, the unusual findings observed in the IFN series are reproduced in a human growth hormone (GH) conjugate series. Subcutaneously infused l20K-GH, GH modified with l-P(EG3Glu), evokes considerably less anti-GH and antipolymer antibodies compared to those modified with dl-P(EG3Glu) or PEG (dl20K-GH or PEG20K-GH). As a result, repeated injections of dl20K-GH or PEG20K-GH, but not l20K-GH, result in a clear ABC effect and significantly diminished drug availability in the blood. Meanwhile, immature mouse bone marrow cells incubated with the helical l20K-GH exhibit decreased drug uptake and secretion of proinflammatory cytokines compared to those treated with one of the other two GH conjugates bearing unstructured polymers. Taken together, the current study highlights an urgent necessity to systematically reassess the pros and cons of choosing unstructured polymers for protein conjugation. Furthermore, our results also lay the foundation for the development of next-generation biohybrid drugs based on helical synthetic polypeptides.
Content may be subject to copyright.
Therapeutic Protein PEPylation: The Helix of Nonfouling Synthetic
Polypeptides Minimizes Antidrug Antibody Generation
Yingqin Hou,
Yu Zhou,
Hao Wang,
Jialing Sun,
Ruijue Wang,
Kai Sheng,
Jingsong Yuan,
Yali Hu,
Yu Chao,
§
Zhuang Liu,
§
and Hua Lu*
,
Beijing National Laboratory for Molecular Sciences, Center for Soft Matter Science and Engineering, Key Laboratory of Polymer
Chemistry and Physics of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing
100871, Peoples Republic of China
Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, Peoples Republic of China
§
Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and
Technology, Soochow University, Suzhou, Jiangsu 215123, China
*
SSupporting Information
ABSTRACT: Polymer conjugation is a clinically proven approach to
generate long acting protein drugs with decreased immune responses.
Although poly(ethylene glycol) (PEG) is one of the most commonly
used conjugation partners due to its unstructured conformation, its
therapeutic application is limited by its poor biodegradability,
propensity to induce an anti-PEG immune response, and the resultant
accelerated blood clearance (ABC) eect. Moreover, the prevailing
preference of unstructured polymers for protein conjugation still lacks
strong animal data support with appropriate control reagents. By using
two biodegradable synthetic polypeptides with similar structural
compositions (L-P(EG3Glu) and DL-P(EG3Glu)) for site-specic
protein modication, in the current study, we systematically
investigate the eect of the polymer conformation on the in vivo pharmacological performances of the resulting conjugates.
Our results reveal that the conjugate L20K-IFN, interferon (IFN) modied with the helical polypeptide L-P(EG3Glu) shows
improved binding anity, in vitro antiproliferative activity, and in vivo ecacy compared to those modied with the
unstructured polypeptide analogue DL-P(EG3Glu) or PEG. Moreover, L20K-IFN triggered signicantly less antidrug and
antipolymer antibodies than the other two. Importantly, the unusual ndings observed in the IFN series are reproduced in a
human growth hormone (GH) conjugate series. Subcutaneously infused L20K-GH, GH modied with L-P(EG3Glu), evokes
considerably less anti-GH and antipolymer antibodies compared to those modied with DL-P(EG3Glu) or PEG (DL20K-GH or
PEG20KGH). As a result, repeated injections of DL20K-GH or PEG20K-GH, but not L20K-GH, result in a clear ABC eect and
signicantly diminished drug availability in the blood. Meanwhile, immature mouse bone marrow cells incubated with the
helical L20K-GH exhibit decreased drug uptake and secretion of proinammatory cytokines compared to those treated with one
of the other two GH conjugates bearing unstructured polymers. Taken together, the current study highlights an urgent necessity
to systematically reassess the pros and cons of choosing unstructured polymers for protein conjugation. Furthermore, our results
also lay the foundation for the development of next-generation biohybrid drugs based on helical synthetic polypeptides.
INTRODUCTION
Therapeutic proteins are important biologics that frequently
exhibit high potency and selectivity. However, their clinical use
has been hampered by their rapid renal clearance, susceptibility
to proteolysis, and strong immunogenicity.
13
Particularly, the
generation of antidrug antibodies (ADAs) has been a serious
hurdle for many protein drugs.
4
One proven strategy to
overcome these limitations is to covalently conjugate the
protein of interest to polymers such as poly(ethylene glycol)
(PEG), a process known as PEGylation, which can lead to
signicantly increased hydrodynamic volume, in vivo stability,
and circulation half-life.
510
However, there is mounting
evidence that PEGylated proteins tend to show poorer binding
anity and biological activity than their unconjugated
equivalents.
11,12
Furthermore, although one of the initial
purposes of PEGylation is for reduced ADA generation, PEG
is known to elicit anti-PEG antibodies that adversely accelerate
the blood clearance of the PEGylated proteins or nano-
particles, known as the ABC eect. As evidence, reduction in
the therapeutic ecacy of many PEGylated proteins, such as
uricase, asparaginase, and interferon (IFN), has been found to
strongly correlate with the occurrence of the anti-PEG immune
response that they induce.
13
More worrisome is the fact that
Received: August 10, 2018
Research Article
http://pubs.acs.org/journal/acscii
Cite This: ACS Cent. Sci. XXXX, XXX, XXXXXX
© XXXX American Chemical Society ADOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
This is an open access article published under an ACS AuthorChoice License, which permits
copying and redistribution of the article or any adaptations for non-commercial purposes.
Downloaded via PEKING UNIV on February 15, 2019 at 12:36:46 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.
the percentage of healthy adults carrying pre-existing anti-PEG
antibodies has increased sharply from 0.2% to 42% during the
past three decades, likely because of their daily exposure to
PEG-containing commodities.
13
Thus, a pressing need in this
eld is seeking new polymers beyond PEGylation.
In recent years, researchers have investigated a wide range of
alternative conjugation partners,
14
including zwitterionic
polymers,
15,16
polyglycerol,
17
glycopolymers,
18
and oligo-
EGylated poly(meth)acrylates,
19,20
with varying degrees of
success. Despite the potential of these methods, the lack of
biodegradability has remained a central problem.
5
Synthetic
polypeptides have been increasingly considered as a biode-
gradable and biocompatible alternative to PEG with great
clinical promise.
2125
There has been evidence that the genetic
fusion of therapeutic proteins/peptides to intrinsically
disordered polypeptides, such as XTEN, PAS, and elastin-like
polypeptides (ELP), can lead to improved pharmacological
performance in vivo.
2632
We envisage that the chemical
modication of proteins by synthetic polypeptides, which we
call PEPylation, could open up enormous possibilities.
3335
Particularly, the chemical diversity of synthetic polypeptides
has been greatly expanded by incorporating noncanonical
amino acids via ring-opening polymerization (ROP) of α-
amino acid N-carboxyanhydrides (NCA) and utilizing D-amino
acids.
20
Notably, during the preparation of this manuscript,
Jiang et al. reported the nonspecic grafting of zwitterionic
polypeptides to uricase,
36
which showed extraordinarily low
immunogenicity and outstanding safety prole in vivo. Their
work underscored the exceptional clinical potential of
PEPylation.
When surveying the aforementioned polymers for protein
modication, one can easily draw the conclusion that
unstructured and exible polymers (e.g., PEG) have long
been the preferred conjugation partners due to their ability to
augment the hydrodynamic volume of the modied protein
and provide an excellent stealth eect that minimizes renal
Scheme 1. Site-Specic Conjugation of Synthetic Polypeptides or PEG To Engineer Therapeutic Proteins (IFN and GH) via
Native Chemical Ligation
Figure 1. Characterization and in vivo pharmacological performances of various IFN conjugates. (A) SDS-PAGE gel, stained by Coomassie blue.
(B) Circular dichroism (CD) spectroscopy. (C) Melting temperature (Tm) measured by thermouoro assay. (D) In vivo pharmacokinetics (i.v.
injection) of wt-IFN (n= 6), L20K-IFN (n= 6), and DL20K-IFN (n= 6), and PEG20KIFN (n= 3). (EF) Tumor growth inhibition curves. BALB/
C-nu mice bearing s.c. OVCAR-3 xenograft (E) or patient-derived xenograft (PDX) tumors (F) were i.v. injected with PBS saline or one of the
IFN-based drugs (n= 7 each); treatments began on day 0, and the black arrows indicate ending of the treatments. The total injection numbers are
six in E and three in F. Data are expressed as mean ±SD. Pvalue is determined by two-way ANOVA (Bonferroni post-test) analysis: *p< 0.05, **p
< 0.01, ***p< 0.001.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
B
ltration and immune attack. Following the same principle,
elementary amino acids are carefully selected in the design of
XTEN to ensure an unstructured conformation and absence of
helical structures.
26
However, it is surprising that there have
been very few studies that attempt to investigate whether the
conformation, particularly the helix, of the polymer has any
eect on the in vivo performance of the protein that it
modies. One practical challenge resides in the diculty of
generating protein conjugates that only dier in the
conformation of the attached polymers to ensure a fair
comparison. We reason that synthetic polypeptides oer an
ideal solution to this problem as their secondary conformations
(e.g., helix and coil) can be easily manipulated by switching the
chirality of the monomers without altering the overall chemical
composition.
37,38
RESULTS
Synthesis and Characterization of Dierent IFN
Polymer Conjugates. Recombinant IFN, an antiviral and
antitumor cytokine, was selected as our rst model drug. For a
fair comparison, we synthesized two chemically similar but
conformationally varied polypeptides (Scheme 1).
34,35
Specif-
ically, monomer γ-(2-(2-(2-methoxyethoxy)ethoxy)ethyl L-
glutamate NCA
39
(L-EG3GluNCA) was polymerized by
trimethylsilyl phenylsulde (PhS-TMS) to yield phenyl
thioester-functionalized L-P(EG3Glu) (Scheme 1). Similarly,
DL-P(EG3Glu) was produced from a racemic mixture of DL-
EG3GluNCA. The molecular weights (MW) of both polymers
were carefully controlled to be 20 kDa, in line with many
clinically approved PEG conjugates. Gel permeation chroma-
tography (GPC) indicated that the two polymers had a similar
MW 2223 kDa and narrow dispersity (D̵) below 1.05
(Figure S1). 1H NMR spectroscopy showed that the two
polymers diered in the chemical shift of the α-H due to the
dierent α-C chirality (Figure S2). As expected, circular
dichroism (CD) spectroscopy revealed that α-helices con-
stituted more than 90% of L-P(EG3Glu), whereas DL-
P(EG3Glu) was unstructured as design (Figure S3). Sub-
sequently, we conjugated each synthetic polypeptide to an IFN
mutant bearing a N-terminal cysteine (Cys-IFN) via native
chemical ligation, thereby forming two PEPylated IFNs
denoted as L20K-IFN and DL20K-IFN (Scheme 1). We also
generated PEG20K-IFN as a positive control by attaching a
thioester-functionalized PEG (MW 20 kDa) to IFN via the
same method (Scheme 1 and Figure S4).
All puried IFN conjugates exhibited a narrow size
distribution based on SDS-PAGE analysis (Figure 1A). L20K-
Table 1. In Vitro Binding, Anti-Proliferative Activity, and in Vivo Pharmacokinetics
a
of wt-IFN and Various IFN Conjugates
sample IC50 (pg/mL) KD(nM) elimination half-life (h)
b
AUC0t((μg/mL)*h)
c
Vd(mL)
d
CI (mL/h)
wt-IFN 8.5 ±1.4 1.0 0.5 ±0.1 0.4 ±0.1 125 ±21.8
L20k-IFN 36.0 ±1.3 5.8 9.6 ±0.6 15.5 ±2.2 167 ±49 3.2 ±0.6
DL2ok-IFN 160 ±4 19.6 7.8 ±0.3 8.6 ±0.7 239 ±49 5.8 ±0.7
PEG20k-IFN 190 ±10 15.9 9.8 ±1.9 17.0 ±3.0 161 ±25 2.9 ±0.4
a
Dose: 50 μg/rat on IFN base.
b
Elimination half-life: Time points used to calculate t1/2βare 312 h (wt IFN), 1272 h (all conjugates).
c
AUC
calculated by logarithmic trapezoidal rule up to 12 h (wt-IFN),72 h (L20K-IFN, DL20K-IFN, and PEG20KIFN).
d
Vdcalculated at 12 h after
intravenous injection.
e
Data are expressed as mean ±SD.
Figure 2. In vivo immune responses triggered by IFN conjugates. (AB) Anti-IFN IgG (A) and IgM (B) contents in the sera measured by ELISA;
the plates were coated with wt-IFN and then incubated with 104-fold (for IgG) or 500-fold (for IgM) prediluted sera in PBS. (CD) Antipolymer
IgG (C) and IgM (D) contents in the sera immunized with various polymer-IFN conjugates; for each polymer-of-interest, the ELISA plates were
coated with the corresponding polymer-GH conjugate. (EF) Antipolymer ELISA assays using free DL-P(EG3Glu) (E) or PEG (F) as the
competition agent; sera immunized with DL20K-IFN or PEG20KIFN (week 4) were prediluted 200-fold and incubated with the corresponding free
polymer at gradient concentrations. Immunization protocol: rats were s.c. infused with L20K-IFN, DL20K-IFN, or PEG20KIFN at a weekly dose 0.2
mg/kg for 4 weeks; sera were drawn from the rats (n= 3) every week starting from week 0. For ELISA analysis, after sera incubation and washing,
all plates were incubated with antimouse IgG-HRP or IgM-HRP, and analyzed by TMB solution (CWBIO). TWEEN was excluded from the
buers in all antipolymer ELISA studies. Data are expressed as mean ±SD Pvalue is determined by two-way ANOVA (Bonferroni post-test)
analysis: *p< 0.05, **p< 0.01, ***p< 0.001.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
C
IFN and DL20K-IFN shared an almost identical apparent MW,
whereas PEG20K-IFN appeared to electrophoresize slightly
slower than its PEPylated counterparts but was still
comparable (Figure 1A). CD spectroscopy suggested that
PEG20K-IFN and DL20K-IFN were similar in helicity as wt-IFN,
whereas L20K-IFN produced a stronger helical signal intensity
(Figure 1B). A thermouoro assay
39
indicated that L20K-IFN
possessed a higher Tm, and therefore greater thermostability,
than both DL20K-IFN and PEG20kIFN (Figure 1C). All
conjugates were shown to be signicantly more resistant to
proteolysis than wt-IFN in trypsin digestion assays (Figure S5).
Surface plasmon resonance (SPR) found the KDvalues for the
binding of L20K-IFN, DL20K-IFN, and PEG20KIFN to human
IFNAR2 were 5.8, 19.6, and 15.9 nM, respectively (Table 1
and Figure S6). Thus, L20K-IFN appeared to be 34 fold
more ecient in its receptor interaction than DL20K-IFN or
PEG20KIFN. Consistently, an in vitro viability assay
demonstrated that the IC50 values of L20K-IFN, DL20K-IFN,
and PEG20KIFN against Daudi cells, an IFN-sensitive human
cancer cell line, were 36, 160, and 190 pg/mL, respectively
(Table 1). This implied that L20K-IFN could induce a
signicantly more potent antitumor eect than DL20K-IFN or
PEG20KIFN does.
In Vivo Pharmacological Performances of IFN
Conjugates. We next measured the pharmacokinetic
parameters of the IFN variants in female SpragueDawley
rats. As shown in Figure 1D and Table 1, the elimination half-
lives (t1/2β)ofL20K-IFN, DL20K-IFN, and PEG20KIFN were
9.6, 7.8, and 9.8 h, respectively, all signicantly longer than the
0.5 h t1/2βof wt-IFN. Interestingly, L20K-IFN was slightly but
consistently longer-lived than DL20K-IFN (Pvalue < 0.05;
reproducible in at least two independent experiments with
dierent batches of materials). This was further evidenced by
the greater AUC of L20K-IFN than that of DL20K-IFN (Table 1).
The in vivo ecacy of the conjugates was further evaluated in
two murine models, one bearing OVCAR-3 tumor xenografts
and the other xenografts derived from a prostate cancer patient
(PDX) (see Materials and Methods). In both cases,
administration of L20K-IFN, which carried the helical L-
P(EG3Glu), led to signicantly slower tumor growth (Figure
1A). The superior antitumor ecacy was further conrmed by
the reduced tumor cell proliferation activity according to Ki-67
staining (Figure S7). No body weight loss was observed in
either model during the treatment with L20K-IFN, suggesting
that the drug was well tolerated under the regimen that we
employed (Figure S8).
Figure 3. In vivo immune responses triggered by GH conjugates. (A) SDS-PAGE gel analysis. (B) Trypsin degradation curves. (CD) Anti-GH
IgG (C) and IgM (D) contents in the sera measured by ELISA; the plates were coated with wt-GH and then incubated with 104-fold (for IgG) or
500-fold (for IgM) prediluted sera in PBS. (EF) Antipolymer IgG (E) and IgM (F) contents in the sera immunized with polymer-GH conjugates;
for each polymer of interest, the ELISA plates were coated with the corresponding polymerIFN conjugate and then incubated with the 200-fold
prediluted sera. Immunization protocol: rats were s.c. infused with L20K-GH, DL20K-GH, or PEG20K-GH at a weekly dose 0.2 mg/kg for 4 weeks; sera
were drawn from the rats (n= 3) every week starting from week 0. For ELISA analysis, after sera incubation and washing, all plates were incubated
with antimouse IgG-HRP or IgM-HRP, and analyzed by TMB solution (CWBIO). TWEEN was excluded from the buers in all antipolymer
ELISA studies. (GI) Blood GH contents at selected time points, measured by ELISA, after the rst and third s.c. injections of L20K-GH (G), DL20K-
GH (H), or PEG20KGH (I). Data are expressed as mean ±SD. Pvalue is determined by two-way ANOVA (Bonferroni post-test) analysis: *p<
0.05, **p< 0.01, ***p< 0.001.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
D
Antibody Generation Triggered by IFN Conjugates.
To investigate the immune response of the conjugates,
SpragueDawley rats were randomly grouped and subcuta-
neously administrated with L20K-IFN, DL20K-IFN, or PEG20K
IFN at a weekly dose of 0.2 mg/kg. Interestingly, sera from the
mice immunized with L20K-IFN showed signicantly lower
levels of anti-IFN IgG and IgM than those receiving DL20K-IFN
or PEG20KIFN (Figure 2AB). Serial dilution of sera from
week 4 revealed that L20K-IFN produced 50100 fold lower
anti-IFN IgG and 510 fold lower IgM titers than those
immunized with DL20K-IFN or PEG20KIFN (Figure S9). In
addition, injection with DL20K-IFN or PEG20KIFN appeared
to also induce a detectable amount of antipolymer antibodies,
particularly IgM (Figure 2CD). The specicity of the
antipolymer antibodies in DL20K-IFN and PEG20KIFN sera
was further validated by the corresponding polymer competi-
tion (Figure 2EF). Strikingly, we discovered that L20K-IFN
exhibited almost no detectable eect on the serum level of
antipolymer IgG or IgM in the immunized rats.
Synthesis of and Immune Responses Triggered by
Dierent GHPolymer Conjugates. To test whether our
ndings observed in the IFN conjugates were also applicable to
other therapeutic proteins, we selected human growth
hormone (GH)
41,42
as our second example and engineered
the protein with a N-terminal cysteine (Cys-GH), similar to
that in Cys-IFN. We next covalently tethered L-P(EG3Glu), DL-
P(EG3Glu), and PEG separately to Cys-GH to generate three
conjugates denoted as L20K-GH, DL20K-GH, and PEG20KGH,
respectively (Scheme 1 and Figure 3A). Trypsin digestion
revealed that L20K-GH was signicantly more resistant to
proteolysis than DL20K-GH and PEG20K-GH (Figure 3B).
Furthermore, injection with L20K-GH provoked substantially
less production of anti-GH IgG and IgM antibodies in rats
from week 2, compared to treatment with DL20K-GH or
PEG20K-GH (Figure 3CD). Serial dilution of sera from week
4 revealed that L20K-GH produced 100 fold lower anti-GH
IgG and 20-fold lower IgM titers than those immunized with
DL20K-GH or PEG20KGH (Figure S10). The same trend was
observed when we measured the levels of antipolymer IgG and
IgM following the immunization (Figure 3EF and Figure
S11). To examine the ABC eect, we measured the blood
concentration of GH at selected time points after the rst and
third injection of each conjugate. The results demonstrated
that infusions of L20K-GH led to very similar blood levels of GH
during the rst 12 h and generated almost no ABC eect in 24
h(Figure 3G, statistically insignicant). In sharp contrast, both
DL20K-GH and PEG20KGH caused a characteristic ABC eect
after the third injection (Figure 3HI). In fact, our ELISA kit
failed to detect blood GH at 24 h following the administration
of DL20K-GH or PEG20KGH (Figure 3HI). As a result, the
AUC024h of L20K-GH were comparable after the rst and third
injection (100% vs 112%), whereas the AUC024h of both
DL20K-GH and PEG20KGH after the third infusion were only
6% of those after the rst drug infusion (Table S1).
BMDC Uptake and Activation. During antibody
production, the antigens are usually internalized, fragmented
Figure 4. BMDC internalization and activation. (A) Flow cytometry analysis of BMDC internalization of various FAM-labeled GH conjugates. (B
D) Flow cytometry analysis of proinammatory cytokines secretion: IL-6 (B), IFN-γ(C), and TNF (D). Freshly separated naï
ve BMDCs were ex
vivo incubated in 24-well plate (5 ×105cells/well) for 6 days and treated with conjugates for 12 h (A) or 24 h (BD) at 37 °C. The cytokines in
the medium were measured with CBA Mouse Inammation kit following manufacturers protocol. The PBS-treated BMDCs were served as
controls.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
E
in lysosome, and displayed on the cell surface by dendritic cells
(DCs) to trigger downstream T cell and B cell response. To
understand the dierent antibody responses triggered by the
conjugates,wesoughttoexaminetheveryrst DC
internalization step. For this, we incubated the GH conjugates
with freshly induced immature mouse bone marrow-derived
dendritic cells (BMDCs), which are widely used for the
assessment of antigen presenting.
43
Flow cytometric analysis
found clear evidence for the internalization of DL20K-GH and
PEG20KGH into BMDCs after 12 h of incubation, whereas
the uptake level of L20K-GH was considerably lower (Figure
4A). Consistently, treatment of BMDCs with L20K-GH resulted
in appreciably less secretion of proinammatory cytokines,
including interleukine-6 (IL-6, Figure 4B), interferon-γ(IFN-γ,
Figure 4C), and tumor necrosis factor (TNF, Figure 4D),
compared to the other two GH conjugates carrying
unstructured polymers.
DISCUSSION
The conjugation of polymers to a protein has been
demonstrated to extend its half-time by increasing its
hydrodynamic volume and mitigating the ADA generation.
1
However, the role that the secondary conformation of a
polymer plays in the resultant protein conjugate has been very
rarely investigated, as unstructured polymers have been the
heavily favored choice in past studies. Notably, the
polypeptideuricase conjugate reported by Jiang focused on
the zwitterionic side chain without studying the secondary
conformation eect.
36
We speculated that peptide-based drugs
and biomaterials covalently modied with α-helical polypep-
tides could exhibit improved proteolytic and thermal stability,
binding, as well as other biological functions over those
conjugated with disordered polymers.
37,4447
To ascertain
whether this is the case, however, one needs to employ
polymers that only dier in conformation. Gratifyingly,
controlled NCA ROP and chemoselective labeling collabo-
ratively enabled us to generate protein conjugates that shared
almost identical modication sites and MWs, and were
attached to nearly the same number of polypeptides with
highly similar chemical compositions.
34
As a result, the
secondary conformation of the tethered polypeptides became
the only major variable. This was corroborated by the GPC
curves of the polymers and the narrow size distributions of the
resultant conjugates on the SDS-PAGE gel (Figure S1,Figures
1A and 3A). Of note, due to the distinct chemical structures of
PEG and our P(EG3-Glu), the migration of those conjugates in
SDS-PAGE gel might not completely correlate their MWs,
which is often observed for other polymer modied proteins.
Our results found the helical polypeptide-bearing L20K-IFN
to have higher binding anity and antiproliferative activity in
vitro than DL20K-IFN and PEG20KIFN, both of which were
attached to unstructured polymers (Table 1). This could be
partially attributed to the less steric hindrance imparted by the
rigid helical polypeptides. Moreover, L20K-IFN exhibited
signicant improvement in circulation half-life and in vivo
ecacy compared to DL20K-IFN (Figure 1D). Taken together,
these data suggested that the conjugation of a rigid helical
polypeptide could improve the blood retention of the modied
protein drug without signicantly aecting its binding anity
or potency, thereby oering a viable solution to the well-
known PEG dilemma.
11
Some of the greatest controversies of PEGylation include the
insucient protection of the conjugated proteins from immune
recognition and the generation of anti-PEG antibodies.
13,48
In
the clinic, the anti-IFN neutralizing antibodies has previously
been observed in nonresponding patients and believed to be
the major reason for their development of resistance.
49
In this
regard, it was remarkable that the administration of L20K-IFN
provoked substantially lower production of anti-IFN, as well as
antipolymer IgG and IgM, than DL20K-IFN or PEG20KIFN
(Figure 2). Importantly, similar results were also obtained from
the GH conjugates, indicating that the benets we observed
were independent of the modied protein (Figure 3). We also
synthesized a left-handed helical polypeptide D-P(EG3Glu)
(23 kDa) and produced two conjugates, D20K-IFN and D20K-
GH (data not shown). We discovered that both D20K-IFN and
D20K-GH, similar to L20K-IFN or L20K-GH, showed almost no
antibody response after repeated administration (data not
shown). The results lent further evidence to the generality of
the helix eect. Moreover, the above study help ruling out the
possibility of D-amino acid-induced antibody production in the
cases of DL20K-IFN and DL20K-GH. Although the exact
mechanistic role of helicity remains insuciently explored, a
number of reasons may count for the unexpected ndings. First
of all, the helical L-P(EG3Glu) seems to provide better
antifouling property than DL-P(EG3Glu) and PEG, and thus
minimizing nonspecic internalization with cells and proteins.
Our initial investigation provided preliminary evidence of
conformation-dependent internalization and activation of
immature BMDCs for those examined proteinpolymer
conjugates. In fact, this helical conformation enhanced
antifouling and anticell adhesion was also observed when the
polypeptides were anchored on gold surfaces.
50
Second, helical
polypeptides are well-known more proteolytic stable (Figure
3B) than those unstructured peptidyl analogues, which may
lead to inecient fragmentation and MHC presentation after
BMDC internalization. More rigorous experimental and
modeling studies are currently ongoing to fully uncover the
molecular mechanism of the unusual helical conformation
eect.
CONCLUSIONS
In conclusion, we generated synthetic polypeptides that only
diered in conformation and compared their eects on the in
vivo therapeutic and immunological properties of the protein
drugs to which they were conjugated. Compared with the
unstructured DL-P(EG3Glu) or PEG, the covalent attachment
of the helical L-P(EG3Glu) to therapeutic proteins (namely,
IFN and GH) led to substantial improvement in a variety of
pharmacological properties, such as binding anity, stability,
and in vivo ecacy. Most interestingly, the helical L-
P(EG3Glu)-conjugated IFN and GH elicited a signicantly
milder immune response and exhibited a much weaker ABC
eect than those modied with unstructured polymers. Thus,
the helical nonfouling polypeptides that we employed could be
excellent alternatives to PEG for mitigating the antibody
response to repeatedly administrated therapeutic proteins,
though whether similar benets apply to more immunogenic
foreign proteins requires further validation. Moreover, our
results suggested that the helical conformation of the synthetic
nonfouling polypeptides played an important role in
minimizing/delaying this antibody response. Taken together,
the current study highlighted an urgent necessity to system-
atically reassess the pros and cons of choosing unstructured
polymers for protein conjugation. Furthermore, our results also
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
F
lay the foundation for the development of next-generation
biohybrid drugs based on helical synthetic polypeptides.
ASSOCIATED CONTENT
*
SSupporting Information
The Supporting Information is available free of charge on the
ACS Publications website at DOI: 10.1021/acscents-
ci.8b00548.
GPC curves, 1H NMR, CD spectra, trypsin degradation
curves, SPR binding curves, Ki-67 stained images of
tumor, relative body weight of mice, antibody titer
curves, AUC024h of GH-polymer conjugates at rst and
third dose (PDF)
AUTHOR INFORMATION
Corresponding Author
*E-mail: chemhualu@pku.edu.cn.
ORCID
Zhuang Liu: 0000-0002-1629-1039
Hua Lu: 0000-0003-2180-3091
Funding
This work was nancially supported by National Key Research
and Development Program of China (2016YFA0201400). We
acknowledge grants from National Natural Science Foundation
of China (21474004 and 21722401). H.L. acknowledges the
startup funding from Youth Thousand-Talents Program of
China.
Notes
The authors declare no competing nancial interest.
ACKNOWLEDGMENTS
We thank Prof. Demin Zhou for sharing the plasmid encoding
the wt-GH, Prof. Wei Wei for the help of BMDC culturing,
and Prof. Richard Lerner, Prof. Xing Chen, Prof. Feng Wang
for helpful discussion.
REFERENCES
(1) Kontos, S.; Hubbell, J. A. Drug development: longer-lived
proteins. Chem. Soc. Rev. 2012,41 (7), 26862695.
(2) Zelikin, A. N.; Ehrhardt, C.; Healy, A. M. Materials and methods
for delivery of biological drugs. Nat. Chem. 2016,8(11), 9971007.
(3) Liu, T.; Du, J. J.; Luo, X. Z.; Schultz, P. G.; Wang, F.
Homogeneously modified immunoglobulin domains for therapeutic
application. Curr. Opin. Chem. Biol. 2015,28,6674.
(4) Krishna, M.; Nadler, S. G. Immunogenicity to biotherapeutics
the role of anti-drug immune Complexes. Front. Immunol. 2016,7, 21.
(5) Pelegri-ODay, E. M.; Lin, E. W.; Maynard, H. D. Therapeutic
protein-polymer conjugates: advancing beyond PEGylation. J. Am.
Chem. Soc. 2014,136 (41), 1432314332.
(6) Cobo, I.; Li, M.; Sumerlin, B. S.; Perrier, S. Smart hybrid
materials by conjugation of responsive polymers to biomacromole-
cules. Nat. Mater. 2015,14 (2), 143159.
(7) White, C. J.; Bode, J. W. PEGylation and dimerization of
expressed proteins under near equimolar conditions with potassium 2-
pyridyl acyltrifluoroborates. ACS Cent. Sci. 2018,4(2), 197206.
(8) Wilson, P.; Nicolas, J.; Haddleton, D. M. Polymerprotein/
peptide bioconjugates. In Chemistry of Organo-Hybrids; John Wiley &
Sons, Inc.: Hoboken, NJ, 2014; p 466.
(9) Dumas, A.; Spicer, C. D.; Gao, Z.; Takehana, T.; Lin, Y. A.;
Yasukohchi, T.; Davis, B. G. Self-liganded Suzuki-Miyaura coupling
for site-selective protein PEGylation. Angew. Chem., Int. Ed. 2013,52
(14), 39163921.
(10) Pasut, G.; Veronese, F. M. State of the art in PEGylation: the
great versatility achieved after forty years of research. J. Controlled
Release 2012,161 (2), 461472.
(11) Fishburn, C. S. The pharmacology of PEGylation: Balancing
PD with PK to generate novel therapeutics. J. Pharm. Sci. 2008,97
(10), 41674183.
(12) Gauthier, M. A.; Klok, H. A. Polymer-protein conjugates: an
enzymatic activity perspective. Polym. Chem. 2010,1(9), 13521373.
(13) Zhang, P.; Sun, F.; Liu, S. J.; Jiang, S. Y. Anti-PEG antibodies in
the clinic: Current issues and beyond PEGylation. J. Controlled Release
2016,244, 184193.
(14) Knop, K.; Hoogenboom, R.; Fischer, D.; Schubert, U. S.
Poly(ethylene glycol) in drug delivery: pros and cons as well as
potential alternatives. Angew. Chem., Int. Ed. 2010,49 (36), 6288
6308.
(15) Liu, S. J.; Jiang, S. Y. Chemical conjugation of zwitterionic
polymers protects immunogenic enzyme and preserves bioactivity
without polymer-specific antibody response. Nano Today 2016,11
(3), 285291.
(16) Keefe, A. J.; Jiang, S. Poly(zwitterionic)protein conjugates offer
increased stability without sacrificing binding affinity or bioactivity.
Nat. Chem. 2012,4(1), 5963.
(17) Frey, H.; Haag, R. Dendritic polyglycerol: a new versatile
biocompatible-material. Rev. Mol. Biotechnol. 2002,90 (34), 257
267.
(18) Mancini, R. J.; Lee, J.; Maynard, H. D. Trehalose glycopolymers
for stabilization of protein conjugates to environmental stressors. J.
Am. Chem. Soc. 2012,134 (20), 84748479.
(19) Liu, M.; Johansen, P.; Zabel, F.; Leroux, J. C.; Gauthier, M. A.
Semi-permeable coatings fabricated from comb-polymers efficiently
protect proteins in vivo. Nat. Commun. 2014,5, 5526.
(20) Gao, W. P.; Liu, W. G.; Mackay, J. A.; Zalutsky, M. R.; Toone,
E. J.; Chilkoti, A. In situ growth of a stoichiometric PEG-like
conjugate at a proteins N-terminus with significantly improved
pharmacokinetics. Proc. Natl. Acad. Sci. U. S. A. 2009,106 (36),
1523115236.
(21) Deming, T. J. Synthesis of side-chain modified polypeptides.
Chem. Rev. 2016,116 (3), 786808.
(22) Talelli, M.; Vicent, M. J. Reduction sensitive poly(l-glutamic
acid) (PGA)-protein conjugates designed for polymer masked-
unmasked protein therapy. Biomacromolecules 2014,15 (11), 4168
4177.
(23) Lu, Y. J.; Mbong, G. N. N.; Liu, P.; Chan, C.; Cai, Z. L.;
Weinrich, D.; Boyle, A. J.; Reilly, R. M.; Winnik, M. A. Synthesis of
polyglutamide-based metal-chelating polymers and their site-specific
conjugation to trastuzumab for auger electron radioimmunotherapy.
Biomacromolecules 2014,15 (6), 20272037.
(24) Song, Z.; Han, Z.; Lv, S.; Chen, C.; Chen, L.; Yin, L.; Cheng, J.
Synthetic polypeptides: from polymer design to supramolecular
assembly and biomedical application. Chem. Soc. Rev. 2017,46
(21), 65706599.
(25) Kramer, J. R.; Onoa, B.; Bustamante, C.; Bertozzi, C. R.
Chemically tunable mucin chimeras assembled on living cells. Proc.
Natl. Acad. Sci. U. S. A. 2015,112 (41), 1257412579.
(26) Schellenberger, V.; Wang, C. W.; Geething, N. C.; Spink, B. J.;
Campbell, A.; To, W.; Scholle, M. D.; Yin, Y.; Yao, Y.; Bogin, O.;
Cleland, J. L.; Silverman, J.; Stemmer, W. P. C. A recombinant
polypeptide extends the in vivo half-life of peptides and proteins in a
tunable manner. Nat. Biotechnol. 2009,27 (12), 11861155.
(27) Hu, J.; Wang, G. L.; Liu, X. Y.; Gao, W. P. Enhancing
pharmacokinetics, tumor accumulation, and antitumor efficacy by
elastin-like polypeptide fusion of interferon alpha. Adv. Mater. 2015,
27 (45), 73207324.
(28) Luginbuhl, K. M.; Schaal, J. L.; Umstead, B.; Mastria, E. M.; Li,
X.; Banskota, S.; Arnold, S.; Feinglos, M.; DAlessio, D.; Chilkoti, A.
One-week glucose control via zero-order release kinetics from an
injectable depot of glucagon-like peptide-1 fused to a thermosensitive
biopolymer. Nat. Biomed. Engin. 2017,1, 0078.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
G
(29) Qin, G. K.; Glassman, M. J.; Lam, C. N.; Chang, D.; Schaible,
E.; Hexemer, A.; Olsen, B. D. Topological effects on globular protein-
ELP fusion block copolymer self-assembly. Adv. Funct. Mater. 2015,
25 (5), 729738.
(30) Petitdemange, R.; Garanger, E.; Bataille, L.; Dieryck, W.;
Bathany, K.; Garbay, B.; Deming, T. J.; Lecommandoux, S. Selective
tuning of elastin-like polypeptide properties via methionine/oxidation.
Biomacromolecules 2017,18 (2), 544550.
(31) Gomes, S.; Leonor, I. B.; Mano, J. F.; Reis, R. L.; Kaplan, D. L.
Natural and genetically engineered proteins for tissue engineering.
Prog. Polym. Sci. 2012,37 (1), 117.
(32) Schlapschy, M.; Binder, U.; Borger, C.; Theobald, I.;
Wachinger, K.; Kisling, S.; Haller, D.; Skerra, A. PASylation: a
biological alternative to PEGylation for extending the plasma half-life
of pharmaceutically active proteins. Protein Eng., Des. Sel. 2013,26
(8), 489501.
(33) Hou, Y.; Zhou, Y.; Wang, H.; Wang, R.; Yuan, J.; Hu, Y.; Sheng,
K.; Feng, J.; Yang, S.; Lu, H. Macrocyclization of interferon-
poly(alpha-amino acid) conjugates significantly improves the tumor
retention, penetration, and antitumor efficacy. J. Am. Chem. Soc. 2018,
140 (3), 11701178.
(34) Yuan, J.; Sun, Y.; Wang, J.; Lu, H. Phenyl trimethylsilyl sulfide-
mediated controlled ring-opening polymerization of alpha-amino acid
N-carboxyanhydrides. Biomacromolecules 2016,17 (3), 891896.
(35) Hou, Y.; Yuan, J.; Zhou, Y.; Yu, J.; Lu, H. A Concise approach
to site-specific topological protein-poly(amino acid) conjugates
enabled by in-situ generated functionalities. J. Am. Chem. Soc. 2016,
138 (34), 1099511000.
(36) Zhang, P.; Jain, P.; Tsao, C.; Yuan, Z.; Li, W.; Li, B.; Wu, K.;
Hung, H. C.; Lin, X.; Jiang, S. Polypeptides with high zwitterion
density for safe and effective therapeutics. Angew. Chem., Int. Ed. 2018,
57 (26), 77437747.
(37) Xiong, M.; Lee, M. W.; Mansbach, R. A.; Song, Z.; Bao, Y.;
Peek, R. M.; Yao, C.; Chen, L. F.; Ferguson, A. L.; Wong, G. C. L.;
Cheng, J. J. Helical antimicrobial polypeptides with radial
amphiphilicity. Proc. Natl. Acad. Sci. U. S. A. 2015,112 (43),
1315513160.
(38) Lu, H.; Wang, J.; Bai, Y.; Lang, J. W.; Liu, S.; Lin, Y.; Cheng, J.
Ionic polypeptides with unusual helical stability. Nat. Commun. 2011,
2, 206.
(39) Chen, C. Y.; Wang, Z. H.; Li, Z. B. Thermoresponsive
polypeptides from pegylated poly-L-glutamates. Biomacromolecules
2011,12 (8), 28592863.
(40) Popp, M. W.; Dougan, S. K.; Chuang, T. Y.; Spooner, E.;
Ploegh, H. L. Sortase-catalyzed transformations that improve the
properties of cytokines. Proc. Natl. Acad. Sci. U. S. A. 2011,108 (8),
31693174.
(41) Wu, L.; Chen, J. X.; Wu, Y. M.; Zhang, B.; Cai, X. C.; Zhang, Z.
W.; Wang, Y.; Si, L. L.; Xu, H.; Zheng, Y. X.; Zhang, C. L.; Liang, C.
G.;Li,J.;Zhang,L.;Zhang,Q.;Zhou,D.M.Preciseand
combinatorial PEGylation generates a low-immunogenic and stable
form of human growth hormone. J. Controlled Release 2017,249,84
93.
(42) Cho, H.; Daniel, T.; Buechler, Y. J.; Litzinger, D. C.; Maio, Z.;
Putnam, A. M.; Kraynov, V. S.; Sim, B. C.; Bussell, S.; Javahishvili, T.;
Kaphle, S.; Viramontes, G.; Ong, M.; Chu, S.; Becky, G. C.; Lieu, R.;
Knudsen, N.; Castiglioni, P.; Norman, T. C.; Axelrod, D. W.;
Hoffman, A. R.; Schultz, P. G.; DiMarchi, R. D.; Kimmel, B. E.
Optimized clinical performance of growth hormone with an expanded
genetic code. Proc. Natl. Acad. Sci. U. S. A. 2011,108 (22), 9060
9065.
(43) Liu, S. Y.; Wei, W.; Yue, H.; Ni, D. Z.; Yue, Z. G.; Wang, S.; Fu,
Q.; Wang, Y. Q.; Ma, G. H.; Su, Z. G. Nanoparticles-based multi-
adjuvant whole cell tumor vaccine for cancer immunotherapy.
Biomaterials 2013,34 (33), 82918300.
(44) Chang, Y. S.; Graves, B.; Guerlavais, V.; Tovar, C.; Packman,
K.; To, K. H.; Olson, K. A.; Kesavan, K.; Gangurde, P.; Mukherjee, A.;
Baker, T.; Darlak, K.; Elkin, C.; Filipovic, Z.; Qureshi, F. Z.; Cai, H.
L.; Berry, P.; Feyfant, E.; Shi, X. G. E.; Horstick, J.; Annis, D. A.;
Manning, A. M.; Fotouhi, N.; Nash, H.; Vassilev, L. T.; Sawyer, T. K.
Stapled alpha-helical peptide drug development: a potent dual
inhibitor of MDM2 and MDMX for p53-dependent cancer therapy.
Proc. Natl. Acad. Sci. U. S. A. 2013,110 (36), E3445E3454.
(45) Moellering, R. E.; Cornejo, M.; Davis, T. N.; Del Bianco, C.;
Aster, J. C.; Blacklow, S. C.; Kung, A. L.; Gilliland, D. G.; Verdine, G.
L.; Bradner, J. E. Direct inhibition of the NOTCH transcription factor
complex. Nature 2009,462 (7270), 182188.
(46) Walensky, L. D.; Bird, G. H. Hydrocarbon-stapled peptides:
principles, practice, and progress. J. Med. Chem. 2014,57 (15), 6275
6288.
(47) Mochida, Y.; Cabral, H.; Miura, Y.; Albertini, F.; Fukushima, S.;
Osada, K.; Nishiyama, N.; Kataoka, K. Bundled assembly of helical
nanostructures in polymeric micelles loaded with platinum drugs
enhancing therapeutic efficiency against pancreatic tumor. ACS Nano
2014,8(7), 67246738.
(48) Kierstead, P. H.; Okochi, H.; Venditto, V. J.; Chuong, T. C.;
Kivimae, S.; Frechet, J. M. J.; Szoka, F. C. The effect of polymer
backbone chemistry on the induction of the accelerated blood
clearance in polymer modified liposomes. J. Controlled Release 2015,
213,19.
(49) van der Eijk, A. A.; Vrolijk, J. M.; Haagmans, B. L. Antibodies
neutralizing peginterferon alfa during retreatment of hepatitis C. N.
Engl. J. Med. 2006,354 (12), 13231324.
(50) Zhang, C.; Yuan, J.; Lu, J.; Hou, Y.; Xiong, W.; Lu, H. From
neutral to zwitterionic poly(alpha-amino acid) nonfouling surfaces:
Effects of helical conformation and anchoring orientation. Biomaterials
2018,178, 728737.
ACS Central Science Research Article
DOI: 10.1021/acscentsci.8b00548
ACS Cent. Sci. XXXX, XXX, XXXXXX
H

Supplementary resource (1)

... PEG alternative polymers include poly(2-oxazoline)s (POX), [2] polysarcosine (PSar), [3] linear polyglycerol (LPG), [4] zwitterionic polymers, [5] poly(methyl acrylate), [6] or polypeptides. [7] The development of protein-polymer conjugates is currently a largely empirical process given by PK considerations such as polymer type and architecture, conjugation chemistry, and thereof, resulting protein-polymer linker structure. [6,8] Moreover, the design of fully active proteinpolymer conjugates required precise anchorage of the polymer chain to the protein surface e.g. ...
Article
Full-text available
Therapeutic proteins are commonly conjugated with polymers to modulate their pharmacokinetics but often lack a description of the polymer‐protein interaction. We deployed limited proteolysis mass spectrometry (LiP‐MS) to reveal the interaction of polyethylene glycol (PEG) and PEG alternative polymers with interferon‐α2a (IFN). Target conjugates were digested with the specific protease trypsin and a “heavy” ¹⁵N‐IFN wild type (IFN‐WT) for time‐resolved quantification of the cleavage dynamics. Interactions between IFN‐α2a and its high‐affinity receptor were detailed by LiP‐MS. Then, 10 kDa polymers of PEG, linear polyglycerol (LPG), and poly(2‐oxazoline) (POX) with two different cyclooctyne linkers (BCN/DBCO) were used for site‐specific bioconjugation to azide functionalized IFN‐α2a. Tryptic events at each cleavage site and in different structural environments (loops/helices) were compared. PEG and LPG were similar, and POX showed a reduced interaction profile with the IFN‐α2a surface. All‐atom molecular dynamics simulations of IFN‐DBCO‐polymer conjugates revealed distinct and transient (below 50 ns) protein‐interaction profiles for PEG, LPG, and POX. Cleavage dynamics of IFN‐polymer conjugates from the BCN handle were homogeneous, pointing to a more conserved IFN structure than DBCO‐polymer conjugates. In summary, time‐resolved LiP‐MS for quantification of cleavage events enhances the structural understanding of transient IFN‐polymer interactions, which may be extended to other bioconjugate types.
... Other types of artificial polypeptide-like polymers have been described with the aim to allow biodegradability, enhance PK properties and reduce immunogenicity versus PEGylated biologics. Examples are the artificial α-helical polypeptide L-P(EG 3 Glu) -based on poly-glutamate whose γcarboxylate groups are modified as (uncharged) esters with short PEG chains -dubbed PEPylation [52] and so-called polypeptides with high zwitterion density such as carboxybetaine-functionalized polypeptides (PepCB) [70]. Both synthetic polymer types can be combined. ...
Article
Introduction: Engineering of the drug half-life in vivo has become an integral part of modern biopharmaceutical development due to the fact that many proteins/peptides with therapeutic potential are quickly cleared by kidney filtration after injection and, thus, circulate only a few hours in humans (or just minutes in mice). Areas covered: Looking at the growing list of clinically approved biologics that have been modified for prolonged activity, and also the plethora of such drugs under preclinical and clinical development, it is evident that not one solution fits all needs, owing to the vastly different structural features and functional properties of the pharmacologically active entities. This article provides an overview of established half-life extension strategies, as well as of emerging novel concepts for extending the in vivo stability of biologicals, and their pros and cons. Expert opinion: Beyond the classical and still dominating technologies for improving drug pharmacokinetics and bioavailability, Fc fusion and PEGylation, various innovative approaches that offer advantages in different respects have entered the clinical stage. While the Fc fusion partner may be gradually superseded by engineered albumin-binding domains, chemical PEGylation may be replaced by biodegradable recombinant amino-acid polymers like PASylation, thus also offering a purely biotechnological manufacturing route.
Article
Full-text available
Hyperuricemia, characterized by elevated uric acid levels, is the primary cause of gout. Recombinant uricase is one of the last‐resort therapies but generates unwanted pro‐inflammatory H2O2 and anti‐uricase antibodies. In this work, we developed an albumin corona‐coated enzyme‐loaded zeolitic imidazolate framework (UCZIF) to sustainably maintain low blood uric acid level without producing H2O2. The corona coating not only preserves loaded enzymes but also reduces macrophage phagocytosis by 73.4% compared to free uricase. In addition, the uptake level of UCZIF by dendritic cells is reduced by 74.1%, and the maturation of dendritic cells is inhibited by 35.4% compared to free uricase. Animal experiments demonstrate that albumin corona‐coated UCZIF effectively lowers blood uric acid level in both acute and diet‐induced chronic hyperuricemia models with significantly increasing the half‐life of uricase. Furthermore, compared to the generation of anti‐uricase antibodies during standalone uricase treatment, the levels of anti‐uricase immunoglobulins are significantly reduced by 65.5% (immunoglobulin M) and 76.3% (immunoglobulin G) with repeated administration of albumin corona‐coated UCZIF. Overall, this albumin corona‐coated nanoscale metal–organic framework offers a promising approach to minimize the immunogenicity induced by exogenous enzymes and further safely reduce uric acid levels in the treatment of hyperuricemia.
Article
Therapeutic proteins are commonly conjugated with polymers to modulate their pharmacokinetics but often lack a description of the polymer‐protein interaction. We deployed limited proteolysis mass spectrometry (LiP‐MS) to reveal the interaction of polyethylene glycol (PEG) and PEG alternative polymers with interferon‐α2a (IFN). Target conjugates were digested with the specific protease trypsin and a “heavy” 15N‐IFN wild type (IFN‐WT) for time‐resolved quantification of the cleavage dynamics. Interactions between IFN‐α2a and its high‐affinity receptor were detailed by LiP‐MS. Then, 10 kDa polymers of PEG, linear polyglycerol (LPG), and poly(2‐oxazoline) (POX) with two different cyclooctyne linkers (BCN/DBCO) were used for site‐specific bioconjugation to azide functionalized IFN‐α2a. Tryptic events at each cleavage site and in different structural environments (loops/helices) were compared. PEG and LPG were similar, and POX showed a reduced interaction profile with the IFN‐α2a surface. All‐atom molecular dynamics simulations of IFN‐DBCO‐polymer conjugates revealed distinct and transient (below 50 ns) protein‐interaction profiles for PEG, LPG, and POX. Cleavage dynamics of IFN‐polymer conjugates from the BCN handle were homogeneous, pointing to a more conserved IFN structure than DBCO‐polymer conjugates. In summary, time‐resolved LiP‐MS for quantification of cleavage events enhances the structural understanding of transient IFN‐polymer interactions, which may be extended to other bioconjugate types.
Article
Full-text available
Lipid nanoparticle (LNP)/mRNA complexes have great therapeutic potential but their PEG chains can induce the production of anti-PEG antibodies. New LNPs that do not contain PEG are greatly needed. We demonstrate here that poly-glutamic acid-ethylene oxide graft copolymers can replace the PEG on LNPs and outperform PEG-LNPs after chronic administration.
Article
Full-text available
A method of poly(ethylene glycol) (PEG) conjugation is known as PEGylation, which has been employed to deliver therapeutic drugs, proteins, or nanoparticles by considering the intrinsic non- or very low immunogenic property of PEG. However, PEG has its weaknesses, and one major concern is the potential immunogenicity of PEGylated proteins. Because of its hydrophilicity, poly(sarcosine) (P(Sar)) may be an attractive—and superior—substitute for PEG. In the present study, we designed a double hydrophilic diblock copolymer, methoxy-PEG-b-P(Sar)m (m = 5–55) (mPEG-P(Sar)m), and synthesized a triblock copolymer with hydrophobic poly(l-isoleucine) (P(Ile)). We validated that double hydrophilic mPEG-P(Sar) block copolymers suppressed the specific binding of three monoclonal anti-PEG antibodies (anti-PEG mAbs) to PEG. The results of our indirect ELISAs indicate that P(Sar) significantly helps to reduce the binding of anti-PEG mAbs to PEG. Importantly, the steady suppression of this binding was made possible, in part, thanks to the maximum number of sarcosine units in the triblock copolymer, as evidenced by sandwich ELISA and biolayer interferometry assay (BLI): the intrinsic hydrophilicity of P(Sar) had a clear supportive effect on PEG. Finally, because we used P(Ile) as a hydrophobic block, PEG-P(Sar) might be an attractive alternative to PEG in the search for protein shields that minimize the immunogenicity of PEGylated proteins.
Article
Full-text available
The covalent conjugation of large, functionalized molecules remains a frontier in synthetic chemistry, as it requires rapid, chemoselective reactions. The potassium acyltrifluoroborate (KAT)-hydroxylamine amide-forming ligation shows promise for conjugations of biomolecules under aqueous, acidic conditions, but the variants reported to date are not suited to ligations at micromolar concentrations. We now report that 2-pyridyl KATs display significantly enhanced ligation kinetics over their aryl counterparts. Following their facile, one-step incorporation onto the termini of polyethylene glycol (PEG) chains, we show that 2-pyridyl KATs can be applied to the construction of protein-polymer conjugates in excellent (>95%) yield. Four distinct expressed, folded proteins equipped with a hydroxylamine could be PEGylated with 2-20 kDa 2-pyridyl mPEG KATs in high yield and with near-equimolar amounts of coupling partners. Furthermore, the use of a bis 2-pyridyl PEG KAT enables the covalent homodimerization of proteins with good conversion. The 2-pyridyl KAT ligation offers an effective alternative to conventional protein-polymer conjugation by operating under aqueous acidic conditions well suited for the handling of folded proteins.
Article
Stealth materials are widely used in nano‐formulations to improve the performance of therapeutics. As the current "gold standard", poly(ethylene glycol) (PEG) polymer effectively promotes the pharmacokinetics of veiled therapeutic cargos while reducing their immune responses. However, recent studies suggested that the usage of PEG could induce adverse reactions, including the emergence of anti‐PEG antibodies and PEG‐induced tissue histologic changes, therefore casting a shadow over its future therapeutic applications. An alternative material with similar or better therapeutic enhancement effect, but no or less immunogenicity and organ toxicity is urgently needed. Herein, we designed a polypeptide with high zwitterion density (PepCB) as a stealth material for therapeutics. Neither any tissue histologic change in liver, kidney and spleen, nor abnormal behavior, sickness or death was induced by the synthesized polymer after a high‐dosage administration regimen for three months in rats. When conjugated to a therapeutic protein uricase, the uricase‐PepCB bioconjugate showed significantly improved pharmacokinetics and immunological properties compared with uricase‐PEG conjugates.
Article
s The development of high-performance nonfouling polymer surfaces for implantable medical devices and therapeutic nanomaterials is of great importance. Elaborating the relationship of polymer structural characteristics and the resulted surface properties can offer useful guidance toward ideal biointerfaces. In this work, we investigate the effects of the helical conformation and anchoring orientation of poly(α-amino acid)s (PαAAs) to produce advanced nonfouling surfaces. By using the neutral poly(γ-(2-(2-(2-methoxyethoxy)ethoxy)ethoxy)esteryl glutamates) (P(EG3Glu)s) as a model system, the adsorption kinetics are monitored by ex-situ variable angle spectroscopic ellipsometry and in-situ quartz crystal microbalance with dissipation. It is found that the polymers adopting a rigid rod-like α-helical conformation can self-assemble more rapidly to produce denser adlayers, and generate significantly improved nonfouling surfaces compared to those flexible polymer analogues including the widely used antifouling polymer PEG. Moreover, the surface properties can be further enhanced by using the antiparallel orientated helical P(EG3Glu)s. Most importantly, the insights gained from the P(EG3Glu) model system are successfully applied to the generation of ultra-low-fouling surfaces using zwitterionic PαAAs brushes, underscoring the generality of the approach. Particularly, the surface based on the antiparallel aligned zwitterionic helical PαAAs exhibits ∼98–99% reduction of human serum adsorption relative to the bare gold, and gives almost no adhesion of mouse platelet. Taken together, this work depicts an extremely simple yet highly effective approach to manipulate surface properties for numerous applications in biomaterial interfaces, diagnostics, and biosensors.
Article
Cyclization and polymer conjugation are two commonly used approaches for enhancing the pharmacological properties of protein drugs. However, cyclization of parental proteins often only affords a modest improvement in biochemical or cell-based in vitro assays. Moreover, very few studies have included a systematic pharmacological evaluation of cyclized protein-based therapeutics in live animals. On the other hand, polymer-conjugated proteins have longer circulation half-lives but usually show poor tumor penetration and suboptimal pharmacodynamics due to increased steric hindrance. We herein report the generation of a head-to-tail interferon-poly(α-amino acid) macrocycle conjugate circ-P(EG3Glu)20-IFN by combining the aforementioned two approaches. We then compared the antitumor pharmacological activity of this macrocycle conjugate against its linear counterparts, N-P(EG3Glu)20-IFN, C-IFN-P(EG3Glu)20 and C-IFN-PEG. Our results found circ-P(EG3Glu)20-IFN to show considerably greater stability, binding affinity, and in vitro antiproliferative activity towards OVCAR3 cells than the three linear conjugates. More importantly, circ-P(EG3Glu)20-IFN exhibited longer circulation half-life, remarkably higher tumor retention and deeper tumor penetration in vivo. As a result, administration of the circular conjugate could effectively inhibit tumor progression and extend survival in mice bearing established xenograft human OVCAR3 or SKOV3 tumors without causing severe paraneoplastic syndromes. Taken together, our study provided till now the most relevant experimental evidence in strong support of the in vivo benefit of macrocyclization of protein-polymer conjugates and for its application in next-generation therapeutics.
Article
Synthetic polypeptides from the ring-opening polymerization of N-carboxyanhydrides (NCAs) are one of the most important biomaterials. The unique features of these synthetic polypeptides, including their chemical diversity of side chains and their ability to form secondary structures, enable their broad applications in the field of gene delivery, drug delivery, bio-imaging, tissue engineering, and antimicrobials. In this review article, we summarize the recent advances in the design of polypeptide-based supramolecular structures, including complexes with nucleic acids, micelles, vesicles, hybrid nanoparticles, and hydrogels. We also highlight the progress in the chemical design of functional polypeptides, which plays a crucial role to manipulate their assembly behaviours and optimize their biomedical performances. Finally, we conclude the review by discussing the future opportunities in this field, including further studies on the secondary structures and cost-effective synthesis of polypeptide materials.
Article
Stimulation of the glucagon-like peptide-1 (GLP1) receptor is a useful treatment strategy for type 2 diabetes due to pleiotropic effects, such as the regulation of islet hormones and the induction of satiety. However, the native ligand for the GLP1 receptor has a short half-life owing to enzymatic inactivation and rapid clearance. Here, we show that a subcutaneous depot formed after a single injection of GLP1 recombinantly fused to a thermosensitive elastin-like polypeptide results in zero-order release kinetics and circulation times of up to 10 days in mice and 17 days in monkeys. The optimized pharmacokinetics lead to 10 days of glycaemic control in three different mouse models of diabetes, as well as the reduction of glycosylated haemoglobin levels and weight gain in ob/ob mice treated once weekly for 8 weeks. Our results suggest that the optimized GLP1 formulation could enhance therapeutic outcomes by eliminating peak-and-valley pharmacokinetics and improving overall safety and tolerability. The design principles that we established should be broadly applicable for improving the pharmacological performance of other peptide and protein therapeutics.
Article
In this study, we aimed to develop a safe and stable form of human growth hormone (hGH) and to refine PEGylation methods for therapeutic proteins via genetic code expansion. Through this precise approach, a series of polyethylene glycol (PEG) moieties and sites were combined in various ways. Additionally, the effects of combinatorial PEGylation on the biological, pharmacological, and immunogenic properties of hGH in vitro and vivo were analyzed. Our results showed that combinatorial PEGylation at Y35, G131, and K145 significantly reduced immunogenicity and improved pharmacokinetic (PK) profiles compared with mono-PEGylation, while retaining biological activity. Upon re-examination of the pharmacodynamics in hypophysectomized rats, multi-PEGylated hGH was found to be much more stable than mono-PEGylated hGH. Thus, this method for combinatorial, precise PEGylation may facilitate the development of next-generation, long-acting hGH with low immunogenicity.
Article
We have designed and prepared a recombinant elastin-like polypeptide (ELP) containing precisely positioned methionine residues, and performed the selective and complete oxidation of its methionine thioether groups to both sulfoxide and sulfone derivatives. Since these oxidation reactions substantially increase methionine residue polarity, they were found to be a useful means to precisely adjust the temperature responsive behavior of ELPs in aqueous solutions. In particular, lower critical solution temperatures were found to be elevated in oxidized sample solutions, but were not eliminated. These transition temperatures were found to be further tunable by the use of solvents containing different Hofmeister salts. Overall, the ability to selectively and fully oxidize methionine residues in ELPs proved to be a convenient post-modification strategy for tuning their transition temperatures in aqueous media.
Article
Biological drugs generated via recombinant techniques are uniquely positioned due to their high potency and high selectivity of action. The major drawback of this class of therapeutics, however, is their poor stability upon oral administration and during subsequent circulation. As a result, biological drugs have very low bioavailability and short therapeutic half-lives. Fortunately, tools of chemistry and biotechnology have been developed into an elaborate arsenal, which can be applied to improve the pharmacokinetics of biological drugs. Depot-type release systems are available to achieve sustained release of drugs over time. Conjugation to synthetic or biological polymers affords long circulating formulations. Administration of biological drugs through non-parenteral routes shows excellent performance and the first products have reached the market. This Review presents the main accomplishments in this field and illustrates the materials and methods behind existing and upcoming successful formulations and delivery strategies for biological drugs. © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.