You are on page 1of 13

Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

Available online at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/AJPS

Original Research Paper

Relaxin-encapsulated polymeric metformin


nanoparticles remodel tumor immune
microenvironment by reducing CAFs for efficient
triple-negative breast cancer immunotherapy

Hongyan Zhang a,b,1, Liying Chen a,1, Yue Zhao a,1, Ningchao Luo a, Jingbin Shi a, Shujun Xu a,
Lisha Ma a, Menglin Wang c, Mancang Gu a, Chaofeng Mu a, Yang Xiong a,b,∗
a School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
b Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
c Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill,

NC 27599, United States

a r t i c l e i n f o a b s t r a c t

Article history: Cancer-associated fibroblasts (CAFs) are one of the most abundant stromal cells in the
Received 25 November 2022 tumor microenvironment which mediate desmoplastic response and are the primary driver
Revised 14 January 2023 for an immunosuppressive microenvironment, leading to the failure of triple-negative
Accepted 9 February 2023 breast cancer (TNBC) immunotherapy. Therefore, depleting CAFs may enhance the effect
Available online 25 February 2023 of immunotherapy (such as PD-L1 antibody). Relaxin (RLN) has been demonstrated to
significantly improve transforming growth factor-β (TGF-β) induced CAFs activation and
Keywords: tumor immunosuppressive microenvironment. However, the short half-life and systemic
Cancer-associated fibroblasts vasodilation of RLN limit its in vivo efficacy. Here, plasmid encoding relaxin (pRLN) to
Plasmid encoding relaxin locally express RLN was delivered with a new positively charged polymer named polymeric
Lipid nanoparticles metformin (PolyMet), which could increase gene transfer efficiency significantly and have
Polymeric metformin low toxicity that have been certified by our lab before. In order to improve the stability of
PD-L1 antibody pRLN in vivo, this complex was further formed lipid poly-γ -glutamic acid (PGA)/PolyMet-
pRLN nanoparticle (LPPR). The particle size of LPPR was 205.5 ± 2.9 nm, and the zeta
potential was +55.4 ± 1.6 mV. LPPR displayed excellent tumor penetrating efficacy and
weaken proliferation of CAFs in 4T1luc /CAFs tumor spheres in vitro. In vivo, it could reverse
aberrantly activated CAFs by decreasing the expression of profibrogenic cytokine and
remove the physical barrier to reshape the tumor stromal microenvironment, which enabled
a 2.2-fold increase in cytotoxic T cell infiltration within the tumor and a decrease in
immunosuppressive cells infiltration. Thus, LPPR was observed retarded tumor growth
by itself in the 4T1 tumor bearing-mouse, and the reshaped immune microenvironment
further led to facilitate antitumor effect when it combined with PD-L1 antibody (aPD-L1).


Corresponding author.
E-mail address: xyxnb@126.com (Y. Xiong).
1
These authors contributed equally to this work.
Peer review under responsibility of Shenyang Pharmaceutical University.

https://doi.org/10.1016/j.ajps.2023.100796
1818-0876/© 2023 Shenyang Pharmaceutical University. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)
2 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

Altogether, this study presented a novel therapeutic approach against tumor stroma using
LPPR to achieve a combination regimen with immune checkpoint blockade therapy against
the desmoplastic TNBC model.
© 2023 Shenyang Pharmaceutical University.
This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/)

key role in the recruitment, activation and infiltration of


1. Introduction regulatory T cells(Treg), macrophages, and myeloid-derived
suppressor cells (MDSC) in TME [30]. Significantly, TGF-β
Breast cancer is the 2nd most prevalent cancer worldwide and activation in CAFs is thought to be a deciding factor of CD8+
the main reason for death in women [1]. Furthermore, triple T cell exclusion and low response to aPD-1/PD-L1 [31,32].
negative breast cancer (TNBC) represents one of the most Therefore, damaging CAFs by TGF-β secretion inhibition may
malignant tumors than other breast cancer subtypes with be promising to facilitate immunotherapy [33–35].
a poorer prognosis, relapse proneness, and drug resistance Relaxin (RLN) is a potent antifibrogen that suppresses
[2,3]. Conventional therapeutic approaches for TNBC are fibroblast activation upon binding to its principal receptor
traditional surgery, chemotherapy, and hormone therapy. RXFP1 (Relaxin family peptide receptor type 1) [33]. Relaxin
However, these therapies display limited therapeutic benefits displays a potent anti-fibrotic based on its specific capacity to
due to molecular and morphological heterogeneity [4,5]. suppress pro-fibrotic cytokine-mediated abnormal fibroblast
Immunotherapy (such as PD-L1 antibody) has shown promise proliferation, differentiation and matrix production [36]. It
in recent years for cancer treatment with a high mutational is well-documented that RLN has a powerful anti-fibrotic
burden, which include melanoma [6], lung [7], and ovarian activity to down-regulate TGF-β-induced collagen production
cancers [8]. Of note, PD-L1 antibody (aPD-L1) provides a long- and contraction of lung, cardiac, renal, and hepatic fibrosis
lasting response rate of up to 40% in melanoma [9]. In [37]. Important to its function, RLN also have maintained
comparison, TNBC possesses elevated PD-L1 expression, but anti-tumor efficacy attributed to its stromal modulation
patients exhibited only a 20% sustained response rate treated function by significantly inhibited TGF-β/pSmad2 signaling
with aPD-L1 immunotherapy [10]. One of the reasons for the pathway [33,38]. Nevertheless, the direct applicability of RLN
treatment failure is the abundant geographic or central tumor was constrained by its relatively small size (6 kDa), potential
fibrosis in TNBC tumor microenvironment (TME) [11,12], off-target effects, short circulating half-life, continuous
which acts as a barrier to immune checkpoint blockade (ICB) administration, and adverse effects such as systemic
therapy [13]. vasodilation [37,39]. Therefore, a feasible approach to delivery
TME is a complex arrangement of tumor cells, reactive of RLN plasmids (pRLN) into tumor tissues by generating local
matrix cells, blood vessels, immune cells, and extracellular expression of RLN may be a prospective strategy.
matrix (ECM), all of which constitute a disorganized and In view of this concern, we introduced the polymeric
aggressive ecological niche compared to well-organized, metformin (PolyMet) to form PolyMet-pRLN complex. PolyMet,
homeostatic healthy tissue [14,15]. As critical effector cells which our lab formulated in previous studies [40–42], was a
that mediate desmoplasia, hyper-proliferative cancer- highly positively charged polymer chain for effective delivery
associated fibroblasts (CAFs) are essential for tumor of genes to beat the bottleneck of “stabilization dilemma” of
progression [16]. Notably, CAFs are also responsible for pDNA delivery [43]. PolyMet satisfies a few standards of an
recruiting immunosuppressive cells, confining cytotoxic T effective gene carrier based on small molecular, excellent
lymphocytes (CTLs), and guarding cancer cells from immune tolerability and low toxicity [44,45]. Several experimental
surveillance [17]. Altogether, this relatively ineffectiveness of studies have provided that pDNA develops a loose and
aPD-L1 therapy might be attributed by the abundance of CAFs non-stable complex when mixed with only polycationic
in TNBC [18–20]. In recent years, nanoparticles have provided polymers owing to its low molecular weight [44] and charge
several smart delivery systems based on active targeting of density [46,47]. Hence, in this work, liposomes were utilized
CAFs and immunotherapy, such as liposomes [21–23] and to develop complexes to develop a novel lipid poly-γ -
polymeric nanoparticles [24,25]. Many efforts have been glutamic acid (PGA)/PolyMet-pRLN nanoparticle (LPPR) to
made both to eradicate CAFs and to reshape their function. guard pDNA from dissociation or removal prior to reaching
Nano-strategies focused on exhausting CAFs are emerging to the tumor. To the end, LPPR, as an in-situ reservoir, could
augment immunotherapy of breast cancer [26]. deliver pRLN to achieve local RLN expression in tumor. LPPR
Among the most prominent characteristics of the dense with a positive charge in physiological conditions displayed
stroma is the excessive expression of transforming growth increased penetrating efficacy and efficient intracellular
factor β (TGF-β) [27,28]. Firstly, under normal conditions, drug release in 4T1luc /CAFs tumor spheres due to pRLN.
normal fibroblasts are typically present in a resting state. We further assessed the antitumor effect of LPPR in 4T1
In TME, they can be stimulated by growth factors of TGF- tumor bearing mouse. LPPR diminished CAFs to change
β to differentiate into CAFs [29]. This activation induces the immunosuppressive environment into an immune
the strengthen of collagen deposition, reshaping ECM, and stimulating state with increased cytotoxic T-cell infiltration.
formation dense stromal TME. Moreover, TGF-β holds a Furthermore, LPPR eventually led to an improvement of aPD-
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 3

Scheme 1 – The proposed mechanism of LPPR in combination with aPD-L1 mediates removal of CAFs and immune cell
infiltration. (A) LPPR formulation. (B) Effective delivery of RLN gene through LPPR with aPD-L1 disrupts the
immunosuppressive and desmoplastic TME by targeting CAFs (a. RLN gene in LPPR could silence abnormally active CAFs; b.
aPD-L1 blocks the binding of PD-1 and PD-L1 and promotes tumor cell killing by CD8+ T cells). (C) LPPR triggers the
eradication or continuous remission of aggressive TNBC through synergy with aPD-L1.

L1 therapeutic efficacy. Our findings suggested the effective staining are listed in Tables S1 and S2 in Supplementary
delivery of the RLN gene through LPPR, as the ideal candidate, Material.
had the dual roles of addressing abundant tumor fibrosis and Cell line: The 4T1luc cells were generously supplied by
immunosuppressive microenvironment, finally facilitated Sichuan University (Sichuan, China). 4T1luc cell line stably
checkpoint blockade in TNBC (Scheme 1). expresses the firefly luciferase gene, which was introduced via
lentiviral transduction.
Experimental animals: Mice (5–7 weeks old, female,
2. Material and methods BALB/c) were acquired from Shanghai SLAC Laboratory
Animal Co., Ltd and bred at the Animal Experimental Research
2.1. Material Center of Zhejiang Chinese Medical University. All animal
studies were conducted in compliance with NIH guidelines for
RLN plasmid was obtained from Sino Biological Inc. Linear laboratory research and were authorized by the Animal Care
PEI hydrochloride (MW = 8000), HOBt, EDC·HCl, and Coumarin Committee of Zhejiang Chinese Medical University. 6 × 105
6 were purchased from Aladdin (Shanghai, China). DOTAP 4T1luc cells were injected orthotopically into the mammary
were bought from Advanced Vehicle Technology (Shanghai, fat pad of BALB/c female mice. Treatment was initiated when
China). BCA protein assay kits and DAPI were obtained the 4T1luc tumor volume reached 100 mm3 in 8–10 d after
from Beyotime (Shanghai, China). CellTracker Green 5- inoculation, the mice were randomly divided into 4 groups
chloromethylfluorescein diacetate (CMFDA) was acquired (n = 5).
from Yeasen Biotechnology Co., Ltd (Shanghai, China). D-
K+ was purchased from PerkinElmer (USA). All the other 2.2. Synthesis of PolyMet
chemicals were sourced from Sigma-Aldrich, except where
otherwise mentioned. PolyMet (MW = 4,300 Da) was synthesized as previously
Antibodies: Anti-mouse PD-L1 (clone 10F.9G2) were described [40,41]. Briefly, PEI (0.4 g) and dicyandiamide (4 g)
obtained from BioXcell (West Lebanon, NH). Antibodies for were initially mixed in 20 ml deionized water. Next, HCl (4 ml)
flow cytometry, Western blotting and immunofluorescence was added and oil bath (100 °C) for 4 h. Then the product was
4 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

Fig. 1 – Physicochemical characterization of LPPR. (A) Agarose gel electrophoresis of PolyMet-pRLN complex. Lane M, DNA
ladder; other lanes, PolyMet-pRLN complex prepared at different N/P ratios. (B) Size and surface charge of
PGA/PolyMet-pRLN at different ratios (1:1, 2:3, 3:2). (C) Size and surface charge of PGA/Lipid at various ratios (1:4, 1:8, 1:16,
1:24). (D) TEM image and size distribution of LPPR (scale bar, 50 nm). (E) Zeta potentials of LPPR. (F) Release profile of pRLN
from LPPR at various pH. Means ± SD (n = 3).

purified through ultrafiltration centrifuge tube (MW = 3500), 2.4. Characterization of LPPR
washed with deionized water for 2 times, and freeze-dried to
obtain PolyMet. The size and surface charge of LPPR were estimated by
ZetaSizer (Malvern ZetaSizer, Malvern, UK). The surface
2.3. Preparation of LPPR morphology images of LPPR were acquired by transmission
electron microscopy (TEM, JEM-2100, JEOL, Japan). The Lipid
The pDNA (pRLN) complexation efficiency with PolyMet was poly-γ -glutamic acid (PGA)/PolyMet nanoparticle (LPP), which
investigated by gel retardation assay with different N/P ratios. was LPPR without loading relaxin plasmids, and coumarin
PolyMet-pRLN complex were induced at various N/P from 6-labeled LPPR nanoparticles were prepared with the same
2 to 8 by adding PolyMet with a particular concentration procedures as LPPR. The amount of pRLN released from LPPR
into pRLN solution. The resulting PolyMet-pRLN complex was confirmed with Hoechst 33,258 nucleic acid staining
was incubated at 25 °C for 15 min. Agarose gels (1%, w/v) (EX = 360 nm, Em = 460 nm) [48].
were prepared containing ethidium bromide (1 mg/ml) in
TAE buffer. PolyMet-pRLN complex were softly blended with 2.5. In vitro release of pRLN from LPPR
loading dye bromophenol blue and xylenol (9:1, v/v) and added
to various wells of the agarose gel. The gel was resolved in TAE The in vitro release profile of pRLN from LPPR was carried out
buffer at 75 V for 20 min, imaged on a UV detector. by dialysis membrane method. In short, 2 ml LPPR solution
Next, PGA was added in distilled water with PolyMet- (0.3 mg pRLN) was put in a dialysis bag (MWCO = 3500) and
pRLN complex (pRLN = 50 μg) in different molar ratios placed in 30 m release medium (pH 7.4 and pH 5.6), then
(2:3, 1:1 and 3:2) to form the core of lipid nanoparticles. shaken at 75 rpm with the temperature of 37 ± 0.2 °C. One
Meanwhile, the lipid membrane of LPPR was prepared by milliliter of release medium at predetermined time points (0.5,
thin-layer dispersion. Briefly, DOTAP (20 mM, 1 ml) and 1, 2, 4, 8, 12, 24, 48, 72 h) were removed and supplemented with
cholesterol (20 mM, 1 ml) were dissolved in chloroform, an equal amount of fresh medium.
and the solvent was eliminated by evaporation to produce
a homogeneous lipid membrane. The lipid membrane 2.6. LPPR penetration by confocal laser scanning
was then hydrated and extruded sequentially through the microscopy in 4T1luc/CAFs tumor spheres
polycarbonate membrane to develop cationic liposomes
(10 mM) (Millipore, Billerica, MA). Finally, to identify the The CAFs and 4T1luc cell were mixed evenly at a ratio of 1:1
optimum amount of liposomes coating on the core, various and placed in ultra-low adsorption 96 well cell plates (6.0 × 104
molar ratios (1:4, 1:8, 1:16, 1:24) of PGA to DOTAP were cells/well) to form an in vitro co-culture mixed tumor model.
investigated. The plates were cultured and photographed daily under an
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 5

Fig. 2 – The effect of LPPR in penetration effect and proliferation of CAFs in 4T1luc /CAFs tumor spheres. (A) Intracellular
distribution and tracing of Coumarin 6-labled LPPR or LPP in 4T1luc /CAFs tumor spheres after 48 h of incubation. (B) Living
cell imaging of CAFs in 4T1luc /CAFs tumor spheres after LPP or LPPR treatment by CLSM (Scale bar=50 nm).

inverted fluorescent microscope to observe morphological 2.7. Effect of LPPR on the proliferation of CAFs in
changes in the tumor spheres.After incubation for 48 h, the 4T1luc/CAFs tumor spheres
4T1luc /CAFs tumor spheres were treated with coumarin 6-
labeled LPP and LPPR with the coumarin-6 concentration at CAFs were incubated with CMFDA (10 μm) for 25 min, and
0.2 μg/ml. After incubation with LPP and LPPR for 48 h, the replaced with fresh medium. The CAFs treated by CMFDA were
tumor spheroids were washed by PBS and then immobilized rinsed with PBS and imaged. Green fluorescence indicated
them with 4% paraformaldehyde (PFA). The cell nucleus was successful CMFDA staining. 4T1luc and CMFDA stained CAFs
dyed with DAPI (5 μg/ml). The tumor spheres were scanned were then used to prepare 4T1luc /CAFs tumor spheres. After
layer-by-layer in “Z-stack” mode by a CLSM (Zeiss, Germany) incubated with LPP and LPPR for 48 h, the tumor spheres were
to observe the penetration of Coumarin 6-labeled LPP and stained with DAPI. The intracellular fluorescent pictures were
LPPR in 4T1luc /CAFs tumor spheres (DAPI: Ex = 358 nm, captured by a CLSM for three-dimensional imaging (CMFDA:
Em = 461 nm; Coumarin 6: Ex = 466 nm, Em = 504 nm). Ex = 492 nm, Em = 517 nm). Finally, the proliferation of CAFs in
6 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

Fig. 3 – LPPR in combination with aPD-L1 therapy on 4T1luc tumor model. (A) Combination regimen of LPPR in combination
with aPD-L1 for antitumor therapy. (B) Tumor growth curves of 4T1luc tumors. (C) The tumor weight. (D) The changes in
tumor luminescence intensity of tumor-bearing mice. (E) Relative BLIrel ratio curve after treatment with different drugs for
different times in vivo. (F) Changes in relative body weight of 4T1luc -bearing mice. (G) The tumor image of various
treatments after 14 d (H) H&E and TUNEL staining of treated 4T1luc tumors. (n = 5, ∗ P < 0.05, ∗∗ P < 0.01).

4T1luc /CAFs tumor spheres was evaluated by the fluorescence The animals were sacrificed 18 d after tumor transplant,
intensity of CMFDA. the tumor, and major organs (heart, liver, spleen, lung, and
kidney) were excised for further H&E and TUNEL staining.
2.8. In vivo antitumor efficacy
2.9. Immune status investigation
Mice bearing 4T1luc allografts were randomly grouped and the
treatments were started on Day 7. Saline, aPD-L1 (5 mg/kg, Infiltrating immune cells including CTLs (CD3+ CD4− CD8+ ), T
ip), LPPR (1.5 mg/kg pRLN, iv) and LPPR + aPD-L1(1.5 mg/kg effector cell (Teff, CD3+ CD4+ CD8− ), Treg (CD3+ CD4+ Foxp3+ ),
pRLN, iv + 5 mg/kg aPD-L1, ip) were administered. d-luciferin MDSC (CD11b+ Gr1+ ), dendritic cell (DC, CD11c+ MHC class
(150 mg/kg) was intraperitoneally injected into anesthetized Ⅱ+ ) and M2 macrophage (CD11b+ F4/80+ CD206+ ) in the TME
mice, and the BLI was documented to evaluate the 4T1luc were profiled by flow cytometry (FCM). Single cell suspensions
growth by an IVIS imaging system every 3 d The body weight acquired first by adding dissociation buffer (200 U/ml
and the tumor volume (Vt ) of mice were calculated every 2 d collagenase I, 200 U/ml collagenase IV, and 100 μg/ml DNase
using Eq. 1. I) to tumors and incubating at 37 °C for 60 min in the shaker.
The single cell suspensions were stained with fluorescently
Vt =(A × B2 )/2 (1) labeled antibodies for analyzing the expression of surface
markers in immune cells (BD FACS LyricTM Flow Cytometer).
Where A was the longest and B was the shortest diameter The results were analyzed via CytExpert software.
of the tumor.
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 7

Fig. 4 – LPPR in combination with aPD-L1 treatment attenuates 4T1luc desmoplastic reaction. (A) Western blot analysis of
FAP-α, TGF-β, α-SMA and GAPDH expression in the 4T1luc tumor model after various therapy. (B) Protein expression levels
are normalized to GAPDH. RT-PCR analysis of (C) TGF-β, FGF2, PDGF-b and PDGF-c expression in the 4T1luc tumor model. (D)
Confocal microscopy is identified α-SMA and CD3+ in tumor tissues after different drug treatments. The quantitative results
of (E) α-SMA and CD3+ are shown as percent (%) of the total cell number. Scale bar represents 100 μm (n = 3, ∗ P < 0.05, ∗∗ P <
0.01 ∗∗∗ P < 0.001).

RT-PCR was carried out for measuring the amount of for 1 h to label CD3 and α-SMA, which were the markers of
interferon gamma (IFN-γ ), tumor necrosis factor-α (TNF-α), T cell and fibrosis in tumor respectively. The cell nuclei were
Interleukin-4 (IL-4), Interleukin-10 (IL-10), TGF-β, fibroblast then stained with DAPI. Images were obtained using a VS120
growth factor 2 (FGF 2), platelet growth factor b and c S6 Slide scanner (Olympus, Japan).
(PDGF-b and PDGF-c). The RNeasy R Microarray Tissue Mini
Kit was utilized to extract RNA from tumor tissue. cDNA 2.10. Western blot
was synthesized and amplified via PCR. Glycerol-3-phosphate
dehydrogenase (GAPDH) was served as the housekeeping To assess the expression of protein in 4T1luc tumors, tumor
control. tissues collected from tumor-bearing mice were lysed using
Tumor tissues collected from tumor-bearing mice were radioimmunoprecipitation assay (RIPA) buffer (containing 1%
immediately placed in 4% PFA for overnight fixation followed protease inhibitor and 1% phosphatase inhibitor), and protein
by paraffin embedding. Afterwards, the slides were soaked concentration was determined using a BCA assay. Samples
in 5% BSA for 15 min after dewaxing, antigen recovery and were diluted and heated at 95 °C for 6 min. Proteins were
permeabilization. The sections were incubated with anti- separated using 10% SDS-polyacrylamide gel electrophoresis
CD3 and anti-alpha smooth muscle actin (α-SMA) primary and then transferred to polyvinylidene difluoride membranes.
antibody overnight at 4 °C, followed by secondary antibody Membranes were blocked with 5% nonfat dry milk in TBST
8 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

and then incubated with primary antibody overnight (4 °C).


Subsequently, the PVDF membranes were incubated with
secondary antibodies and the bands were detected using
the Pierce ECL Western Blotting substrate (Bio-Rad) for
chemiluminescent detection. ImageJ was utilized to quantify
the relative levels of protein expression with GAPDH as an
internal control.

2.11. In vivo safety evaluation

On Day 18 after tumor transplant, the serum was separated.


Creatinine (CREA), blood urea nitrogen (BUN), serum alanine
aminotransferase (ALT), and aspartate transaminase (AST)
were detected the hepatorenal functions. Major organs
consisting of heart, liver, lung, kidney, and spleen were
gathered, and immobilized for H&E staining to assess organ-
specific toxicity.

2.12. Statistical analysis

Statistical results were presented as mean ± SD. The statistical


significance was estimated by one-way ANOVA or two-tailed
Fig. 5 – LPPR in combination with aPD-L1 changed
t-test, where a P value < 0.05 was deemed statistically
cytokines in the TME using quantitative RT-PCR.
significant.
Intra-tumoral changes of (A) Th1 and (B) Th2 cytokines.
n = 3, ∗ P < 0.05, ∗∗ P < 0.01 ∗∗∗ P < 0.001.

Fig. 6 – LPPR in combination with aPD-L1 therapy remodeled the TIME on 4T1luc tumor model. (A-B) FCM analysis of CD8+
(gated on CD3+ ), (C) CD4+ T cells (gated on CD3+ ), (D) Treg, (E) Activated DCs, (F) MDSC and (G-H) M2-Macrophage in the
4T1luc tumors (n = 3). ∗ P < 0.05, ∗∗ P< 0.01, ∗∗∗ P < 0.001.
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 9

of nanoparticles by damaging the CAFs in the tumor


3. Results and discussion sphere.

3.1. Preparation and of characterization LPPR


3.4. Antitumor efficacy of LPPR in combination with
aPD-L1 therapy in 4T1 bearing mice
PolyMet (MW = 4,300 Da) was synthesized via a one-step
reaction (Fig. S1A) [40,41]. Since the fully protonated state of
Having demonstrated that LPPR was able to damaging the
guanidine at physiological pH, the highly positively charged
CAFs in vitro, we next examined whether this tactic enhanced
PolyMet could be utilized to condensate pRLN. Electrophoresis
the tumor inhibitory effect of aPD-L1 in the TNBC. Firstly,
assays (Fig. 1A) suggested PolyMet thoroughly condense pRLN
orthotopic 4T1luc bearing mice were treated in accordance
at a mass ratio above 4:1. PGA with PolyMet-pRLN in a ratio
with the timeline illustrated in Fig. 3A. As compared with PBS
of 3:2 resulted in the smallest core (173.6 ± 0.3 nm) with
group, monotherapies of LPPR exhibited a restricted efficacy
a zeta potential of −45.5 ± 2.0 mV (Fig. 1B). The molar
in inhibiting the growth of 4T1luc tumors. Unsurprisingly, 4T1
ratio of PGA to DOTAP was selected at 1ࢼ16 to provide an
tumors with low levels of immunogenicity and strong levels
appropriate positive charge and a favorable size to maintain
of immunosuppression did not react to αPD-L1. Notably, the
its stability in vivo (Fig. 1C). The average particle size of
combination of LPPR with aPD-L1 showed improved antitumor
LPPR was 205.5 ± 2.9 nm. TEM images showed the spherical
properties, suggesting sufficient capacity to sensitize TNBC to
morphology of LPPR (Fig. 1D). An increase in zeta potential
aPD-L1 in combination with LPPR (Fig. 3B and 3C). No body
from −45.5 ± 2.0 mV for the PGA/(PolyMet-pRLN) complex to
weight loss was detected in the treatment period, supporting
+55.4 ± 1.6 mV for LPPR indicated successful coating of the
the security of the combination therapy (Fig. 3D). In vivo BLI
cationic lipid bilayer on the core (Fig. 1E). The stabilization of
imaging and quantification analysis of 4T1luc -bearing mouse
LPPR in a serum-containing medium was studied. The particle
receiving therapies also demonstrated that LPPR + aPD-L1
size of LPPR was maintained at 215 ± 10 nm and the PDI was
group could prevent tumor progression during the period of
always between 0.1–0.2 over 48 h (Fig. S2). From these data, it
treatment in accordance with the tumor images (Fig. 3E–3G).
could be concluded that LPPR has excellent physical stability
A further assessment of the antitumor efficacy of LPPR was
in serum-containing media.
performed by using H&E and TUNEL staining to study the
apoptosis of 4T1luc cells. All groups displayed various levels
3.2. In vitro release of pRLN from LPPR
of tumor inhibition, with LPPR + aPD-L1 generated maximal
tumor apoptosis (Fig. 3H).
To estimate the DNA plasmid release profile of LPPR in vitro,
pRLN in LPPR was released in PBS at pH 7.4 and pH 5.6. The
3.5. LPPR reshaped the stromal microenvironment
pRLN released from LPPR showed an initial slow release at
first, followed by a continuous fast release step, and finally
Accumulating experimental evidence demonstrated
reached a stable release. The initial slow release was lasted for
the importance of intra-tumoral T-cell infiltration for
8 h, with approximately 10% and 15% of pRLN released from
the antitumor efficacy of aPD-L1 immunotherapy [52].
LPPR at pH 7.4 and 5.6. The continuous fast release profile of
Unfortunately, CAFs engage in tumor-tumor biologic
LPPR until 48 h reached a steady state, in which nearly 35%
interactions by secreting various cell factors and ECM
and 60% of pRLN release at pH 7.4 and 5.6. At last, a negligible
proteins, such as α-SMA and FAP-α to facilitate tumor
pRLN release was observed after 48 h. These results showed
proliferation and limit penetration of T cells [53,54]. Moreover,
that pRLN release from LPPR in vitro demonstrated a sustained
TGF-β was the most potent and ubiquitous profibrogenic
release profile.
cytokine promoting CAFs activation and ECM deposition [11].
Therefore, after three consecutive dosing regimens, tumors
3.3. LPPR decreased the proliferation of CAFs and thus
were analyzed by western blot to assess α-SMA, FAP-α, and
facilitated its penetration into 4T1luc/CAFs tumor spheres
TGF-β (Fig. 4A). All pRLN-containing treatments (LPPR group
and LPPR + aPD-L1 group) clearly reduced TGF-β, FAP-α
The in vitro 4T1luc /CAFs tumor spheres preserved a matrix and
and α-SMA, signaling the damage of CAFs. LPPR + aPD-L1
three-dimensional (3D) structure resembling the TME in vivo
achieved a reduction in α-SMA (>50%), FAP (>40%) and TGF-β
[49,50]. CAFs, as the predominant stromal cells in TME, play
(>30%) compared with PBS (Fig. 4B). RT-PCR demonstrated
a vital role in the restricted penetration of drug in the tumor
that TGF-β was reduced 2.7-fold by LPPR + aPD-L1 treatment
tissue [26,51]. Here, the in vitro CAFs proliferation caused by
(P<0.001). Other pro-fibroblastic growth cytokines which
LPPR was evaluated by the fluorescence intensity of CMFDA. It
participate in the facilitation of collagen synthesis and
was found that LPPR could damage CAFs more efficiency than
suppression of extracellular matrix degradation such as FGF
LPP (Fig. 2B).
2, PDGF-b and PDGF-c [55], also displayed markedly reduced
To observe the penetration ability of LPPR after acting
levels in comparison to the PBS group (Fig. 4C). Likewise,
on CAFs, Coumarin 6 was used as a fluorescent probe in
the LPPR group markedly alleviated the tumor inhibition by
the encapsulated liposomes. The results demonstrated that
significantly diminishing fibrosis (α-SMA expression) and
Coumarin 6-labeled LPP was located on the border of the
recruiting CD3+ T cells in IHC (Fig. 4D–4E). These results
tumor, while coumarin 6-labeled LPPR could penetrate deeper
suggested that the combination of LPPR with αPD-L1 could
regions of the tumor (Fig. 2A). All these results demonstrated
diminish the stromal barrier within TNBC due to the presence
that pRLN could effectively increase the penetration
of LPPR.
10 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

Fig. 7 – Toxicity evaluation for LPPR in combination with aPD-L1 therapy. (A) Serum ALT, (B) AST, (C) BUN, and (D) CREA
levels. (E) H&E staining of major organs after different treatments. Scale bar = 200 μm, n = 3.

3.6. LPPR remodels the immune status to improve activated CAFs should be responsible for recruiting circulating
aPD-L1 therapy in 4T1 bearing mice myeloid cells and Treg. A significantly reduced number of
immunosuppressive Treg was noted in the LPPR-treated group
As we demonstrated LPPR have impact on damaging CAFs, (Fig. 6D). With the combination of LPPR and aPD-L1, the
the relationship between therapy and immune response number of Treg was reduced from 7.96% in PBS group to 4.03%.
was evaluated further. Firstly, we detected the variations We further investigated the DCs population in TME by FCM.
in immune cytokines in the TME. We discovered elevated Activated DCs were significantly higher in each treatment
relative mRNA expression of TNF-α and IFN-γ after treatment group compared to the PBS, and the combination group
with LPPR + aPD-L1 (Fig. 5A) [56]. In contrast, the RLN gene displayed the largest population of DC cells (Fig. 6E). Upon the
therapy substantially downregulated T helper cell 2 (Th2) stimulation by the suppressive Th2 cytokines (e.g., IL-4 and IL-
cytokine signal including IL-4 and IL-10 (Fig. 5B). Notably, IL- 10), these infiltrated myeloid cells differentiated into MDSC
4 has a profibrotic function by promoting the differentiation and M2 macrophages [61]. It has been shown that recruited
of quiescent resident fibroblast to myofibroblast [57]. Upon MDSC and M2 macrophages were negatively correlated with
stimulation with IL-4 and IL-10, the recruited myeloid cells the amount of CTLs in the TME, leading to poor prognosis
will differentiate toward immunosuppressive phenotypes in TNBC (Fig. 6F–6H). These observations demonstrated the
(DC, M2 macrophage, and MDSC, etc.) [58]. Together, these capability of combined infiltration enhancement of CTLs by
immunosuppressive factors block T cell proliferation and LPPR and aPD-L1 in TNBC immunotherapy. Therefore, we
differentiation, thereby provoke the death of functional CTLs suggested LPPR, which apparently reversed the resistance
and facilitate tumor cell evasion from immune surveillance mechanism of aPD-L1 treatment by damaging the CAFs,
[59]. stimulating the anti-tumor immunity and unleashing the
T-infiltrating lymphocytes, especially CD8+ cytotoxic T ability of aPD-L1 to destroy tumors.
cell-mediated cellular immunity is the primary mechanism
of antitumor immunity [60]. As expected, deactivation of 3.7. Biosafety evaluation
CAFs by LPPR significantly reduced intra-tumoral IL-4 and IL-
10 (Fig. 5B), which resulted in the increased infiltration of The safety of LPPR + aPD-L1 therapy was further investigated
CD3+ (1.3-fold increase), CD8+ (2.2-fold increase) and CD4+ T in vivo. BALB/c mice orthotopically bearing 4T1luc were
cells (1.5-fold increase) into TME compared to PBS (Figs. S3 administered with PBS, aPD-L1, LPPR, and LPPR + aPD-L1. No
and 6A–6C). Apart from directly impairing T cell function, obvious weight reductions were observed during the various
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 11

treatments (Fig. 4E). Additionally, we gathered peripheral demonstrated that delivery the plasmid encoding RLN
blood for serum biochemical analysis. No abnormal variation by PolyMet damaged CAFs and tumor growth as well as
in serum ALT, AST, BUN, or CREA were detected in each potentiated the effect of aPD-L1 in TNBC. LPPR with a positive
treatment group, indicating high liver and kidney safety of charge in physiological conditions displayed increased
the regimen (Fig. 7A-7D). Finally, H&E staining showed that no tumor penetrating efficacy and weaken proliferation of
histological abnormalities in the heart, liver, lungs, spleen, and CAFs in 4T1luc /CAFs tumor spheres to overcome the stromal
kidneys in any treatments (Fig. 7E). The above results indicated microenvironment in vitro. Additionally, LPPR markedly
through the whole treatment, no adverse hematological or enhanced the tumor immune microenvironment (TIME)
histological toxicity were observed from LPPR combined with and therapeutic efficiency of aPD-L1 by promoting fibrosis
aPD-L1. regression and CD8+ T cell infiltration in the mouse model of
TNBC, allowing the immunosuppressive environment shifted
to an immunostimulatory state. Furthermore, this LPPR-
3.8. Discussion mediated combined therapy with aPD-L1 displayed significant
anti-tumor potentials in vivo and excellent biosafety. Taken
Accumulating evidence shows that CAFs are key factors together, we present LPPR as a combination strategy based
in the restricted penetration of drug in the tumor tissue. on antifibrotic and ICB therapy which can be used as a
CAFs contribute to the immunosuppressive cells in TME by powerful TME regulator to achieve enhanced antitumor
secreting proinflammatory cytokines and chemokines [62,63]. potential.
Owing to the importance of intra-tumoral T-cell infiltration,
αPD-L1 could achieve the optimum antitumor efficacy.
Unfortunately, the stromal barrier and immunosuppressive Conflicts of interest
milieu limited penetration of T cells into the TME, which
led to unresponsiveness to αPD-L1 immunotherapy. Not The authors have declared that no competing interest exists.
surprisingly, the TME containing CAFs drastically limits the
promises of effective immunotherapeutic and checkpoint
inhibitors for the treatment of TNBC. Accordingly, it is Acknowledgments
appealing to design a multifunctional therapy that is capable
of exhausting CAFs and reshaping TME. We gratefully acknowledge the efforts Professor Leaf Huang
RLN has been shown to be efficacious in inhibiting or and his group in synthesizing PolyMet. We appreciate
reversing fibrosis in cardiac and renal fibrosis models by TGF- the great experimental support from the Public Platform
β/pSmad2 signaling pathway [33]. But, concerns regarding of Medical Research Center, Academy of Chinese Medical
enzymatic degradation and rapid elimination are often raised Science, Zhejiang Chinese Medical University. This work was
in its application. To overcome the drawbacks of RLN, the funded by the Medical and Health Science and Technology
plasmid DNA encoding RLN was used to locally express RLN Program of Zhejiang Province (2021KY813), the National
in the TME. But, how to effectively deliver pRLN to tumor Natural Science Foundation of China (82174095), and the
sites to remove CAFs has become a critical issue. In the National Natural Science Foundation of Zhejiang Province
present study, we novelty introduced PolyMet, which our lab (LZ22H290001).
formulated in previous studies, to deliver pRLN effectively
to form LPPR nanoparticles. The advantage of PolyMet was
multifold including the highly positively charged potential Supplementary materials
and synthesize easily. Of note, there have been no studies
on developing carrier by PolyMet to delivery pDNA and Supplementary material associated with this article can be
combination with aPD-L1 on TNBC. found, in the online version, at doi:10.1016/j.ajps.2023.100796.
In this study, we provided a therapeutic idea by LPPR that
loaded with antifibrotic pRLN was tested in murine TNBC.
references
The pRLN could damage CAFs and decrease the stromal
deposition to recruit CTLs. In an alternative perspective, the
anti-fibrosis therapy could broad the engagement between
[1] Feigelson HS, Bodelon C, Powers JD, Curtis RE, Buist DS,
αPD-L1 and tumor cells. Through the whole treatment, no
Veiga LH, et al. Body mass index and risk of second
adverse hematological or histological toxicity were observed cancer among women with breast cancer. J Natl Cancer Inst
from LPPR combined with αPD-L1. As a result, the combination 2021;113(9):1156–60.
of checkpoint inhibitors with LPPR should enhance its efficacy [2] Yin L, Duan JJ, Bian XW, Yu SC. Triple-negative breast cancer
in TNBC. molecular subtyping and treatment progress. Breast Cancer
Res 2020;22(1):1–13.
[3] Da Silva JL, Nunes NCC, Izetti P, De Mesquita GG, De
Melo AC. Triple negative breast cancer: a thorough review of
4. Conclusion
biomarkers. Crit Rev Oncol Hematol 2020;145:102855.
[4] Jain V, Kumar H, Anod HV, Chand P, Gupta NV, Dey S,
The abnormally dense fibrosis network in TNBC formed a et al. A review of nanotechnology-based approaches for breast
therapeutic barrier that hinders penetration and antitumor cancer and triple-negative breast cancer. J Control Release
immunotherapy. In this study, we for the first time 2020;326:628–47.
12 Asian Journal of Pharmaceutical Sciences 18 (2023) 100796

[5] Engebraaten O, Vollan HKM, Børresen-Dale AL. cells and cancer-associated fibroblasts by dual functional
Triple-negative breast cancer and the need for new peptide-modified liposomes. Acta Biomater 2019;99:339–49.
therapeutic targets. Am J Pathol 2013;183(4):1064–74. [23] Chen B, Dai W, Mei D, Liu T, Li S, He B, et al. Comprehensively
[6] Auslander N, Zhang G, Lee JS, Frederick DT, Miao B, priming the tumor microenvironment by cancer-associated
Moll T, et al. Robust prediction of response to immune fibroblast-targeted liposomes for combined therapy with
checkpoint blockade therapy in metastatic melanoma. Nat cancer cell-targeted chemotherapeutic drug delivery system.
Med 2018;24(10):1545–9. J Control Release 2016;241:68–80.
[7] Veatch JR, Simon S, Riddell SR. Tumor-infiltrating lymphocytes [24] Guo J, Zeng H, Chen Y. Emerging nano drug delivery
make inroads in non-small-cell lung cancer. Mol Hum Reprod systems targeting cancer-associated fibroblasts for improved
2021(8):27. antitumor effect and tumor drug penetration. Mol. Pharm.
[8] Gjoerup O, Brown CA, Ross JS, Huang RSP, Schrock A, 2020;17(4):1028–48.
Creeden J, et al. Identification and utilization of biomarkers [25] Zheng S, Wang J, Ding N, Chen W, Chen H, Xue M, et al. Prodrug
to predict response to immune checkpoint inhibitors. Aaps J polymeric micelles integrating cancer-associated fibroblasts
2020;22(6):132. deactivation and synergistic chemotherapy for gastric cancer.
[9] Zhao XD, Subramanian S. Intrinsic resistance of solid J Nanobiotechnol 2021;19(1):1–18.
tumors to immune checkpoint blockade therapy. Cancer Res [26] Hu J, Yuan X, Wang F, Gao H, Liu X, Zhang W. The progress
2017;77(4):817–22. and perspective of strategies to improve tumor penetration
[10] Denkert C, Liedtke C, Tutt A, Von Minckwitz G. Molecular of nanomedicines. Chin Chem Lett 2021;32(4):1341–7.
alterations in triple-negative breast cancer-the road to new [27] Karamitopoulou E. Tumour microenvironment of pancreatic
treatment strategies. Lancet 2017;389(10087):2430–42. cancer: immune landscape is dictated by molecular and
[11] Xu H, Hu MY, Liu MR, An S, Guan KY, Wang ML, histopathological features. Brit J Cancer 2019;121(1):5–14.
et al. Nano-puerarin regulates tumor microenvironment [28] Pickup M, Novitskiy S, Moses HL. The roles of TGFβ
and facilitates chemo- and immunotherapy in murine triple in the tumour microenvironment. Nat Rev Cancer
negative breast cancer model. Biomaterials 2020:235. 2013;13(11):788–99.
[12] Cheng Y, Sun R, He M, Zhang M, Hou X, Sun Y, [29] Jena BC, Sarkar S, Rout L, Mandal M. The transformation of
et al. Light-switchable diphtherin transgene system cancer-associated fibroblasts: current perspectives on the role
combined with losartan for triple negtative breast cancer of TGF-β in CAF mediated tumor progression and therapeutic
therapy based on nano drug delivery system. Int J Pharmaceut resistance. Cancer Lett 2021;520:222–32.
2022;618:121613. [30] Truffi M, Mazzucchelli S, Bonizzi A, Sorrentino L,
[13] Hu C, He X, Chen Y, Yang X, Qin L, Lei T, et al. Metformin Allevi R, Vanna R, et al. Nano-strategies to target breast
mediated PD-L1 downregulation in combination with cancer-associated fibroblasts: rearranging the tumor
photodynamic-immunotherapy for treatment of breast microenvironment to achieve antitumor efficacy. Int J
cancer. Adv Funct Mater 2021;31(11):2007149. Mol Sci 2019;20(6):1263.
[14] Costa A, Kieffer Y, Scholer-Dahirel A, Pelon F, [31] Zhao F, Evans K, Xiao C, Devito N, Theivanthiran B,
Bourachot B, Cardon M, et al. Fibroblast heterogeneity Holtzhausen A, et al. Stromal fibroblasts mediate anti–PD-1
and immunosuppressive environment in human breast resistance via MMP-9 and dictate TGFβ inhibitor sequencing
cancer. Cancer Cell 2018;33(3):463 -+. in melanoma. Cancer Immunol Res 2018;6(12):1459–71.
[15] Wen SY, Hou YX, Fu LX, Xi L, Yang D, Zhao MJ, [32] Tauriello DV, Palomo-Ponce S, Stork D, Berenguer-Llergo A,
et al. Cancer-associated fibroblast (CAF)-derived IL32 Badia-Ramentol J, Iglesias M, et al. TGFβ drives immune
promotes breast cancer cell invasion and metastasis evasion in genetically reconstituted colon cancer metastasis.
via integrin beta 3-p38 MAPK signalling. Cancer Lett Nature 2018;554(7693):538–43.
2019;442:320–32. [33] Mardhian DF, Storm G, Bansal R, Prakash J. Nano-targeted
[16] Kalluri R. The biology and function of fibroblasts in cancer. relaxin impairs fibrosis and tumor growth in pancreatic
Nat Rev Cancer 2016;16(9):582–98. cancer and improves the efficacy of gemcitabine in vivo. J
[17] Zhang YL, Li Y, Chen K, Qian L, Wang P. Oncolytic virotherapy Control Release 2018;290:1–10.
reverses the immunosuppressive tumor microenvironment [34] Liu M, Song W, Huang L. Drug delivery systems targeting
and its potential in combination with immunotherapy. tumor-associated fibroblasts for cancer immunotherapy.
Cancer Cell Int 2021;21(1):262. Cancer Lett 2019;448:31–9.
[18] Panagi M, Voutouri C, Mpekris F, Papageorgis P, Martin MR, [35] Chen Y, Hu M, Wang S, Wang Q, Lu H, Wang F,
Martin JD, et al. TGF-β inhibition combined with cytotoxic et al. Nano-delivery of salvianolic acid B induces the
nanomedicine normalizes triple negative breast cancer quiescence of tumor-associated fibroblasts via interfering
microenvironment towards anti-tumor immunity. with TGF-β1/Smad signaling to facilitate chemo-and
Theranostics 2020;10(4):1910. immunotherapy in desmoplastic tumor. Int J Pharmaceut
[19] Desbois M, Wang Y. Cancer-associated fibroblasts: key players 2022;623:121953.
in shaping the tumor immune microenvironment. Immunol [36] Shi XK, Young CD, Zhou HM, Wang XJ. Transforming
Rev 2021;302(1):241–58. growth factor-beta signaling in fibrotic diseases and
[20] Li Z, Shi H, Xie H, Yang Y, Zheng Y, Chen H, cancer-associated fibroblasts. Biomolecules 2020;10(12).
et al. Tri-component programmable nanoregulator with [37] Samuel C, Royce S, Hewitson T, Denton K, Cooney T,
Three-pronged penetration boosts immunotherapy of Bennett RG. Anti-fibrotic actions of relaxin. Br J Pharmacol
Triple-Negative breast cancer. Chem Eng J 2022;439:135712. 2017;174(10):962–76.
[21] Li C, Li Z, Gong X, Liu J, Zheng T, Wang F, et al. Acidic [38] Rizvi S, Gores GJ. The two faces of relaxin in cancer: antitumor
tumor microenvironment-sensitive liposomes enhance or protumor? Hepatology 2020;71(3):1117–19.
colorectal cancer therapy by acting on both tumor [39] Hu MY, Wang Y, Xu LG, An S, Tang Y, Zhou XF, et al. Relaxin
cells and cancer-associated fibroblasts. Nanoscale gene delivery mitigates liver metastasis and synergizes with
2021;13(23):10509–25. check point therapy. Nat Commun 2019;10.
[22] Chen X, Yu Q, Liu Y, Sheng Q, Shi K, Wang Y, et al. Synergistic [40] Xiong Y, Zhao Y, Miao L, Lin CM, Huang L. Co-delivery
cytotoxicity and co-autophagy inhibition in pancreatic tumor of polymeric metformin and cisplatin by self-assembled
Asian Journal of Pharmaceutical Sciences 18 (2023) 100796 13

core-membrane nanoparticles to treat non-small cell lung [51] Pei Y, Chen L, Huang Y, Wang J, Feng J, Xu M,
cancer. J Control Release 2016;244:63–73 Pt A. et al. Sequential targeting TGF-β signaling and KRAS mutation
[41] Zhao Y, Wang W, Guo S, Wang Y, Miao L, Xiong Y, increases therapeutic efficacy in pancreatic cancer. Small
et al. PolyMetformin combines carrier and anticancer 2019;15(24):1900631.
activities for in vivo siRNA delivery. Nat Commun 2016;7:11822. [52] Doroshow DB, Bhalla S, Beasley MB, Sholl LM, Kerr KM,
[42] Xiong Y, Song WT, Shen LM, Wang Y, Zhang J, Hu MY, Gnjatic S, et al. PD-L1 as a biomarker of response
et al. Oral metformin and polymetformin reprogram to immune-checkpoint inhibitors. Nat Rev Clin Oncol
immunosuppressive microenvironment and boost immune 2021;18(6):345–62.
checkpoint inhibitor therapy in colorectal cancer. Adv Ther [53] Kawase T, Yasui Y, Nishina S, Hara Y, Yanatori I, Tomiyama Y,
2020;3(12):2000168. et al. Fibroblast activation protein-alpha-expressing
[43] Jia L, Gao Y, Zhou T, Zhao XL, Hu HY, Chen DW, et al. Enhanced fibroblasts promote the progression of pancreatic ductal
response to PD-L1 silencing by modulation of TME via adenocarcinoma. BMC Gastroenterol 2015;15.
balancing glucose metabolism and robust co-delivery [54] Yu QW, Qiu Y, Li JP, Tang X, Wang XH, Cun XL, et al. Targeting
of siRNA/Resveratrol with dual-responsive polyplexes. cancer-associated fibroblasts by dual-responsive
Biomaterials 2021;271:120711. lipid-albumin nanoparticles to enhance drug perfusion for
[44] Algarni A, Pilkington EH, Suys EJ, Al-Wassiti H, Pouton CW, pancreatic tumor therapy. J Control Release 2020;321:564–75.
Truong NP. In vivo delivery of plasmid DNA by lipid [55] Zhang ZQ, Liu X, Shen ZL, Quan J, Lin CW, Li XR,
nanoparticles: the influence of ionizable cationic et al. Endostatin in fibrosis and as a potential candidate of
lipids on organ-selective gene expression. Biomater Sci anti-fibrotic therapy. Drug Deliv 2021;28(1):2051–61.
2022;10(11):2940–52. [56] Mahlangu T, Dludla PV, Nyambuya TM, Mxinwa V,
[45] Shy BR, Vykunta VS, Ha A, Talbot A, Roth TL, Nguyen DN, Mazibuko-Mbeje SE, Cirilli I, et al. A systematic review
et al. High-yield genome engineering in primary cells using a on the functional role of Th1/Th2 cytokines in type 2 diabetes
hybrid ssDNA repair template and small-molecule cocktails. and related metabolic complications. Cytokine 2020:126.
Nat Biotechnol 2022:1–11. [57] Villarreal-Leal RA, Healey GD, Corradetti B. Biomimetic
[46] Santhanes D, Wilkins A, Zhang H, Aitken RJ, Liang M. immunomodulation strategies for effective tissue repair and
Microfluidic formulation of lipid/polymer hybrid restoration. Adv. Drug Deliv. Rev. 2021;179:113913.
nanoparticles for plasmid DNA (pDNA) delivery. Int J [58] Qian BZ, Pollard JW. Macrophage diversity enhances tumor
Pharmaceut 2022;627:122223. progression and metastasis. Cell 2010;141(1):39–51.
[47] Huang Y, Ding X, Qi Y, Yu B, Xu FJ. Reduction-responsive [59] Jiang H, Hegde S, Denardo DG. Tumor-associated fibrosis
multifunctional hyperbranched polyaminoglycosides with as a regulator of tumor immunity and response to
excellent antibacterial activity, biocompatibility and gene immunotherapy. Cancer Immunol Immun 2017;66(8):1037–48.
transfection capability. Biomaterials 2016;106:134–43. [60] Han C, Kwon BS. Chimeric antigen receptor T-cell therapy for
[48] Huang YK, Chen Y, Zhou SL, Chen L, Wang JH, Pei YY, cancer: a basic research-oriented perspective. Immunother
et al. Dual-mechanism based CTLs infiltration enhancement -Uk 2018;10(3):221–34.
initiated by Nano-sapper potentiates immunotherapy against [61] Ito S, Shirota H, Kasahara Y, Saijo K, Ishioka C. IL-4
immune-excluded tumors. Nat Commun 2020;11(1):622. blockade alters the tumor microenvironment and augments
[49] Wallstabe L, Göttlich C, Nelke LC, Kühnemundt J, Schwarz T, the response to cancer immunotherapy in a mouse model.
Nerreter T, et al. ROR1-CAR T cells are effective against lung Cancer Immunol Immun 2017;66(11):1485–96.
and breast cancer in advanced microphysiologic 3D tumor [62] Raman D, Baugher PJ, Thu YM, Richmond A. Role of
models. JCI insight 2019;4(18):e126345. chemokines in tumor growth. Cancer Lett 2007;256(2):137–65.
[50] Monteiro CF, Custódio CA, Mano JF. Bioengineering a [63] Kitamura T, Qian BZ, Pollard JW. Immune cell promotion of
humanized 3D tri-culture osteosarcoma model to assess metastasis. Nat Rev Immunol 2015;15(2):73–86.
tumor invasiveness and therapy response. Acta Biomater
2021;134:204–14.

You might also like