ArticlePDF AvailableLiterature Review

Gene Therapy Using Nanocarriers for Pancreatic Ductal Adenocarcinoma: Applications and Challenges in Cancer Therapeutics

Authors:
  • Seoul St. Mary's Hospital, The Catholic University of Korea

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide, and its incidence is increasing. PDAC often shows resistance to several therapeutic modalities and a higher recurrence rate after surgical treatment in the early localized stage. Combination chemo-therapy in advanced pancreatic cancer has minimal impact on overall survival. RNA interference (RNAi) is a promising tool for regulating target genes to achieve sequence-specific gene silencing. Here, we summarize RNAi-based therapeutics using nanomedicine-based delivery systems that are currently being tested in clinical trials and are being developed for the treatment of PDAC. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) genome editing has been widely used for the development of cancer models as a genetic screening tool for the identification and validation of therapeutic targets, as well as for potential cancer therapeutics. This review discusses current advances in CRISPR/Cas9 technology and its application to PDAC research. Continued progress in understanding the PDAC tumor microen-vironment and nanomedicine-based gene therapy will improve the clinical outcomes of patients with PDAC.
Pharmaceutics 2022, 14, 137. https://doi.org/10.3390/pharmaceutics14010137 www.mdpi.com/journal/pharmaceutics
Review
Gene Therapy Using Nanocarriers for Pancreatic Ductal
Adenocarcinoma: Applications and Challenges in Cancer
Therapeutics
Eun-Jeong Won 1, Hyeji Park 2, Tae-Jong Yoon 1 and Young-Seok Cho 2,*
1 Laboratory of NanoPharmacy, College of Pharmacy, Research Institute of Pharmaceutical Science and
Technology (RIPST), Ajou University, 206 Worldcup-ro, Yeongtong-gu, Suwon 16499, Korea;
won4456@naver.com (E.-J.W.); tjyoon@ajou.ac.kr (T.-J.Y.)
2 Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of
Medicine, The Catholic University of Korea, Seoul 06591, Korea; phj8637@naver.com
* Correspondence: yscho@catholic.ac.kr; Tel.: +82-1-2258-6021
Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide,
and its incidence is increasing. PDAC often shows resistance to several therapeutic modalities and
a higher recurrence rate after surgical treatment in the early localized stage. Combination chemo-
therapy in advanced pancreatic cancer has minimal impact on overall survival. RNA interference
(RNAi) is a promising tool for regulating target genes to achieve sequence-specific gene silencing.
Here, we summarize RNAi-based therapeutics using nanomedicine-based delivery systems that
are currently being tested in clinical trials and are being developed for the treatment of PDAC.
Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9
(Cas9) genome editing has been widely used for the development of cancer models as a genetic
screening tool for the identification and validation of therapeutic targets, as well as for potential
cancer therapeutics. This review discusses current advances in CRISPR/Cas9 technology and its
application to PDAC research. Continued progress in understanding the PDAC tumor microen-
vironment and nanomedicine-based gene therapy will improve the clinical outcomes of patients
with PDAC.
Keywords: pancreatic ductal adenocarcinoma; miRNA; siRNA; CRISPR/Cas9; nanocarrier
1. Introduction
Pancreatic ductal adenocarcinoma (PDAC) is the third leading cause of cancer re-
lated deaths in the Unites States and the seventh leading cause worldwide. Its incidence
is increasing by 0.5% to 1.0% per year, and it is thought it will be the second leading
cause of cancer-related death by 2030 [1]. As its early diagnosis is very difficult, ap-
proximately 80% of patients present with locally advanced or metastatic disease [2].
PDAC often shows resistance to several therapeutic modalities and a higher recurrence
rate after surgical treatment [3]. Adjuvant chemotherapy with 5-fluorouracil, leucovorin,
irinotecan, and oxaliplatin (FOLFIRINOX), without bolus fluorouracil (modified
FOLFIRINOX) leads to a median overall survival (OS) of 54.4 months among patients
with resected pancreatic cancer [4]. However, combination chemotherapy such as
FOLFIRINOX or nab-paclitaxel plus gemcitabine in advanced pancreatic cancer has
minimal impact on OS in the range of weeks to months [5,6]. In addition, a number of
clinical trials have tested pathway-specific targeted therapeutic agents, such as vascular
endothelial growth factor inhibitors and multi-kinase inhibitors, alone or combined with
conventional chemotherapy in metastatic pancreatic cancer but failed to demonstrate
clinical meaningful benefits [7]. Although immune checkpoint inhibitors, such as cyto-
Citation: Won, E.-J.; Park, H.; Yoon,
T.-J.; Cho, Y.-S. Gene Therapy Using
Nanocarriers for Pancreatic Ductal
Adenocarcinoma: Applications and
Challenges in Cancer Therapeutics.
Pharmaceutics 2022, 14, 137.
https://doi.org/10.3390/
pharmaceutics14010137
Academic Editors: Elena
Mourelatou, Sophia Hatziantoniou,
Eleftheria Galatou and Gabriele
Grassi
Received: 20 November 2021
Accepted: 31 December 2021
Published: 6 January 2022
Publisher’s Note: MDPI stays neu-
tral with regard to jurisdictional
claims in published maps and insti-
tutional affiliations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license
(https://creativecommons.org/license
s/by/4.0/).
Pharmaceutics 2022, 14, 137 2 of 16
toxic T lymphocyte protein 4 (CTLA4) and programmed cell death protein 1 (PD-1) have
shown promise for the treatment of multiple cancers, these agents demonstrate limited
responses for the treatment of patients with PDAC, probably due to the multiple im-
mune-regulatory pathways within the pancreatic tumor microenvironment (TME) [8].
At present, the initiating mechanisms of PDAC are relatively well understood. Most
PDACs develop from premalignant lesions called pancreatic intraepithelial neoplasias
(PanINs), which progress stepwise from low grade to high grade in types 1, 2, and 3.
Subsequently, they progress to invasive lesions with the accumulation of various genetic
alterations. Approximately 90% of PanINs of all grades have point mutations in the
KRAS oncogene (particularly within codon 12). In addition, mutational inactivation of
tumor suppressor genes, including cyclin-dependent kinase inhibitor 2A, tumor protein
53 (TP53), and SMAD family member 4 (SMAD4), are frequently detected in type 2 and 3
lesions. These findings suggest that KRAS mutations are associated with tumor initiation,
and subsequent gene mutations are a rate-limiting step for tumor progression [2,9].
However, the molecular mechanisms underlying metastatic spread still need to be clari-
fied. The TME of PDAC is heterogeneous and characterized by dense stroma, which
consists of proliferating myofibroblasts (pancreatic stellate cells), extracellular matrix
proteins and tumor vasculature, and inflammatory cells, including macrophages, mast
cells, plasma cells, and lymphocytes [10]. Extensive desmoplastic stroma and severe
hypovascularity of PDAC make it difficult to effectively deliver therapeutic agents to
PDAC cells and are associated with poor prognosis and increased tumor invasion and
metastatic spread [11]. A number of therapeutic agents to target or modify the TME are
currently being evaluated but have shown poor and contradictory results due to the
multi-faceted nature of tumor stroma. Genomic profiling has made novel therapeutics
feasible for a small subset of patients, but current approaches require further testing in
larger clinical trials [12].
Nanotechnology has led to significant advances in the diagnosis and treatment of
various malignancies. To successfully treat PDAC, it requires effective delivery of drugs
into the TME as well as tumor cells. Delivery using nanomaterial-based carrier systems
enhances the antitumor activity of conventional chemotherapeutic agents including
gemcitabine, fluorouracil, doxorubicin, and paclitaxel. The development and application
of these drugs for the treatment of pancreatic cancer have been comprehensively re-
viewed elsewhere [13]. In addition, there have been great advances in the development of
efficient delivery systems for gene therapy for the cancer treatment. Non-viral gene de-
livery systems are highly effective and are safer and easier to synthesize than delivery
systems using viral vectors. For successful gene therapy, a number of delivery systems
including liposomes, polymers, and inorganic nanoparticles have been developed and
investigated with cancer-targeting moieties [14].
In this review, we present RNA interference (RNAi)-based therapeutics using na-
nomedicine-based delivery systems that are currently being investigated in clinical trials
for the treatment of PDAC. Recently, we developed a novel therapeutic strategy that
targets KRAS and TP53 mutations at same time via liposome delivery of clustered regu-
larly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9
(Cas9), ribonucleoprotein (RNP) complexes, and single-guide RNA (sgRNA) to over-
come drug-resistance of PDAC, and this treatment significantly enhanced the anti-tumor
activity of gemcitabine [15]. In addition, current advances in the application of genome
editing technology to PDAC research are reviewed. Finally, we provide our perspective
on the development of gene therapy using nanotechnology for future clinical translation.
2. RNAi Therapy for Pancreatic Cancer
RNAi is a process that regulates target genes using double-stranded RNAs (dsR-
NAs), which bind to sequences complementary to a gene’s coding sequence, resulting in
the degradation of corresponding mRNAs and subsequent inhibition of translation to
proteins. It can be achieved using three major different types of RNAi molecules: small
Pharmaceutics 2022, 14, 137 3 of 16
interfering RNA (siRNA), microRNA (miRNA), and short hairpin RNA (shRNA). RNAi
is initiated by Dicer enzyme mediated processing of dsRNA to smaller fragments (~22
nucleotides) of siRNA, which are then incorporated into RNA-induced silencing com-
plexes [16]. Gene therapy using RNAi therapeutics is promising and improves precise
gene delivery to treat human diseases including various cancers. Patisiran is an siRNA
enveloped in lipid NPs, which inhibits the hepatic synthesis of transthyretin (TTR) and
improves several clinical manifestations of hereditary TTR-mediated amyloidosis (HTA)
[17]. It was approved in 2018 as the first RNAi-based therapy by the United States Food
and Drug Administration and European Union for the treatment of HTA in adults [18].
However, there are several limitations of RNAi-based therapies, including easy nucle-
ase-induce degradation within body fluids such as serum and inefficient delivery to de-
sired cells, tissues, and organs. To overcome these problems, various nanocarriers have
been developed, and these are highly attractive since they are safer and easier to synthe-
size than delivery systems using viral vectors. They fall into two categories: organic
complexes (lipid complexes, conjugated polymers, and cationic polymers) and inorganic
NPs (magnetic NPs, quantum dots, carbon nanotubes, and gold NPs).
2.1. RNAi Using miRNAs
miRNAs are 1825 nucleotides in length and are endogenous non-coding RNA
molecules that bind, at least partially, to complementary mRNA sequences, subsequently
inducing target mRNA cleavage and degradation. Unlike siRNA, miRNAs can regulate
multiple mRNAs rather than just one. miRNAs play significant roles in the expression of
genes involved in cancer initiation, growth, progression, metastasis, drug resistance, and
therapeutic efficacy. In addition, those secreted by exosomes from cancer cells regulate
intercellular communication processes in the TME [19]. In PDAC, some miRNAs aber-
rantly express and regulate cancer initiation, progression, and invasion.
miR-21 functions as an oncogene and plays significant roles in cancer cell prolifera-
tion, differentiation, and survival in addition to cancer initiation and progression, which
suggests that it may be a promising therapeutic target for PDAC [20]. miR-21, miR-196a,
miR-196b, and let-7i are highly expressed and strongly associated with low survival in
patients with PDAC [21,22]. Researchers have developed anti-miR-21 oligonucleo-
tide-loaded tumor-penetrating complexes that consist of a C-terminal cell-penetrating
peptide iRGD and an N-terminal fatty acid group to enhance hydrophobic interactions
for the self-assembly of NPs (TPN-21). TPN-21 inhibits miR-21 expression and PDAC
growth in PDO and upregulates phosphatase and tensin homolog and programmed cell
death 4, resulting in strong suppression of tumor growth. Li et al. developed polyeth-
ylene glycol-polyethylenimine-magnetic iron oxide NPs conjugated with anti-CD44v6
single chain variable fragments for co-delivery of miR-21 antisense oligonucleotides and
gemcitabine into PDAC cells [23]. These NPs induce apoptosis and inhibit tumor growth
and metastasis in vitro and in vivo, suggesting synergistic anti-tumor effects of miR-21
gene silencing and chemotherapy in PDAC.
Xie et al. developed a local delivery system combining the silencing of miR-210,
KRASG12D, and blockade of C-X-C motif chemokine receptor 4 (CXCR4) with cholesterol-
modified polymeric CXCR4 antagonist (PCX) NPs for the co-delivery of anti-miR-210
and siKRASG12D [24]. Cholesterol is conjugated to improve the efficacy of enhanced per-
meability and retention (EPR)-independent delivery. PCX NPs block cancer-stroma in-
teractions, anti-miR-210 inactivates stroma-producing pancreatic stellate cells, and
siKRASG12D kills pancreatic cancer cells. In this study, NPs were delivered via intraperi-
toneal (IP) administration to an orthotopic PDAC mouse model as an effective
EPR-independent approach for targeting peritoneal tumors. There was nearly 15-fold
higher tumor accumulation of NPs with local IP delivery compared to intravenous (IV)
delivery.
Wu et al. revealed that miR-9 is positively associated with doxorubicin (DOX) sen-
sitivity and developed supramolecular NPs condensed with an arginine-based plectin-1
Pharmaceutics 2022, 14, 137 4 of 16
(PL-1)-targeting chimeric peptide to bind RNA and target PDAC. PL-1/miR-9 NPs sig-
nificantly enhanced DOX efficacy in PDAC cells and tumor growth from PDXs, which
suggests that miR-9/eIF5A2 might be a novel potential drug for the synergistic therapy of
PDAC [25].
miRNA-150 is a tumor suppressor that is downregulated in the majority of human
pancreatic cancer tissues, which suggests that restoration of miR-150 might be a thera-
peutic strategy for pancreatic cancer [26]. Arora et al. developed poly
(D,L-lactide-co-glycolide) (PLGA)-based nanoformulation (NF) for the delivery of miR-150
to pancreatic cancer cells, and treatment with it resulted in growth, clonogenicity, motility,
and invasion via significant downregulation of target gene mucin 4 in vitro [27].
Although several preclinical studies have demonstrated the efficacy of miRNA-based
gene therapy for the treatment of PDAC, many attempts at developing miRNA therapeu-
tics have not been successful in clinical trials [28]. In addition, clinical trials for miR-
NA-based therapeutics have not been performed for the treatment of PDAC to date.
2.2. RNAi Using siRNAs
siRNA is a short non-coding ds-RNA of 2123 nucleotides, which can induce RNAi
silencing without eliciting non-specific interferon after introduction into the mammalian
cells. It has the potential to silence genes encoding proteins that cannot be controlled by
small molecules and programmable drugs [29]. Non-viral delivery systems using NPs
enhance accumulation in tumor cells via EPR resulting from several factors including
escape from the immune system, improved target to tumor and cell uptake, and pro-
longed circulation time [30]. Over the past two decades, a number of NP-based siRNA
therapeutics have been developed and are being investigated in clinical trials.[31] In this
section, recent advances in RNAi therapeutics using siRNA are described, focusing on
siRNAs currently in clinical trials (Table 1).
Table 1. Nanocarriers-based siRNA therapeutics in clinical trials for the treatment of pancreatic
cancer.
Therapeutic siRNAs
Indication
Target gene
/Protein
Route of
administrations
Phase/Status
Reference
CALLA-01
Cancer, Solid tumors
RPM2
Systemc/IV
I/Terminated
[32]
Atu027
Pancreatic cancer
PKN3
Systemc/IV
II/Completed
[33,34]
siG12D LODER
Pancreatic cancer
KRASG12D
mutation
Local/Surgical
implantation
II/Recruiting
[35]
TKM 080301
Solid tumors with liver
involvement
PLK-1
Systemc/IV
II/Completed
[36]
iExosomes
Pancreatic cancer
KRASG12D
mutation
Systemc/IV
I/Recruiting
[37]
NBF 006
Non-small cell lung,
Colorectal, and Pancreatic
GSTP
Systemc/IV
I/Recruiting
[38]
Abbreviation: GSTP, Glutathione-S-transferase; IV, intravenous; PKN2, protein kinase N3; PLK1,
polo-like kinase 1; RPM2, ribonuclease reductase M2; SNALP, Stable nucleic acid lipid particle.
Approximately 90% of PDAC patients have a KRAS mutation, which plays a major
role of PDAC initiation. Kamerkar et al. developed exosomes derived from normal fi-
broblast mesenchymal cells carrying siRNA or shRNA for silencing KRASG12D [37]. IP
injection of these engineered exosomes inhibited tumor growth and increased survival
compared to liposomes due to their CD47-mediated protection from phagocytosis by
macrophages and monocytes. A Phase I trial of patients with metastatic pancreatic cancer
with KRASG12D mutation is underway (NCT03608631). Glutathione-S-transferase
(GSTP) is a phase II detoxifying enzyme related to cell integrity maintenance, oxidative
Pharmaceutics 2022, 14, 137 5 of 16
stress, and protection against DNA damage. It is a regulator of proteins involved in RAS
signaling pathways and is highly expressed in several cancers with KRAS mutation in-
cluding lung, colorectal, and pancreatic cancers [39]. NBF-006, a lyophilized lipid NP,
consists of an ionizable, non-immunogenic, biodegradable lipid enveloping GSTP siR-
NA. Treatment with NBF-006 significantly inhibits tumor growth in KRAS mutant
non-small cell lung cancer (NSCLC) xenograft models and prolongs survival in surgically
implanted orthotopic lung tumor mice without toxicity [38]. It has recently entered a
Phase I trial for previously treated NSCLC with KRAS mutation and previously treated
progressive or metastatic NSCLC, pancreatic, and colorectal cancer (NCT03819387).
Khvalevsky et al. developed the first local prolonged siRNA delivery system (called
the Local Drug EluteR, or LODER), which is a miniature biodegradable polymeric matrix
that releases siRNA over a period of few months after administration to the tumors [40].
Treatment with LODER-encapsulated anti-KRASG12D siRNA (siG12D LODER) inhibits
cancer cell proliferation and epithelial-mesenchymal transition with significant decrease
in KRAS levels and inhibits orthotopic pancreatic tumor growth and prolonged mouse
survival in vivo. In a Phase 1/2a clinical trial that included 15 patients with inoperable
PDAC, treatment with standard of care chemotherapy following siG12D LODER inser-
tion into the tumor with a needle during an endoscopic ultrasound biopsy procedure
resulted in stable disease in 10 patients and a partial response in 2 patients with favorable
safety data (NCT01188785) [35]. In an ongoing trial that started in 2017, 80 patients with
unresectable or borderline resectable locally advanced pancreatic cancer have been as-
signed to receive repeated doses of 2.8 mg siG12D LODER with chemotherapy (gem-
citabine and nab paclitaxel or FOLFIRINOX) or chemotherapy alone (NCT01676259).
Ribonuclease reductase M2 subunit (RPM2) is correlated with biological behaviors of
tumor cells including proliferation, invasion, migration, cell cycle, and apoptosis and plays
an important role in tumorigenesis in several cancer types [41]. CALLA-01, a targeted NP
system, consists of cyclodextrin-containing polymer, polyethylene glycol (PEG), human
transferrin as a targeting ligand for binding transferrin receptors, and siRNA designed to
reduce expression of RPM2 for tumor inhibition and/or tumor size reduction. In 2008, it
entered the first human Phase I trial involving systemic administration of siRNA to pa-
tients with solid cancer including PDAC (NCT00689065). These NPs were successfully
delivered into cancer cells, and it was confirmed by cancer cells containing nanoparticles
in tumor biopsies from patients with melanoma after systemic administration. In addi-
tion, the expression of RPM2 mRNA and protein is reduced in patients who receive the
highest doses, which suggests that specific gene inhibition by an RNAi might be one
cancer treatment modality [42]. In a Phase Ia/Ib trial, CALLA-01 showed similar safety
profile in 24 patients with different cancers compared to animals [32].
Atu027 is an RNAi therapeutic formulation based on cationic lipids. It contains
neutral fusogenic, PEG-modified lipid components and siRNA molecules that specifi-
cally target protein kinase N3 in the vascular endothelium, a downstream effector of the
phosphoinositol-3-kinase signaling pathway [43]. Atu027 inhibits tumor growth and
lymph node metastasis in orthotopic mouse models for prostate and pancreatic cancer
mouse models and hematogenous metastasis in mouse models for spontaneous lung
cancer [43,44]. In the first human Phase I clinical trial of this compound (NCT01808638),
10 escalating doses of Atu027, as a single IV administration, followed by twice weekly
doses for a 28-day cycle, were given to 24 patients with advanced solid tumors. The
treatment was safe and resulted in tumor stabilization in 41% of patients. In addition,
most patients had a reduced soluble variant of vascular endothelial growth factor re-
ceptor-1, which suggests its potential as a biomarker [33]. In a Phase II trial, 23 patients
with metastatic pancreatic cancer received combination treatment of gemcitabine and
Atu027 (NCT01808638). The treatment was safe, and twice weekly Atu027 led to signifi-
cantly improved progression-free survival [34].
Polo-like kinase 1 (PLK1), an essential cell-cycle protein, plays multiple roles in mi-
tosis and cytokinesis. It is overexpressed in various types of cancer and negatively affects
Pharmaceutics 2022, 14, 137 6 of 16
patient outcome. In addition, inhibition of PLK1 expression induces mitotic arrest and
apoptosis, resulting in tumor growth inhibition [45]. Stable nucleic acid lipid particles
(SNALPs) are effective siRNA delivery system, and do not need active targeting moieties
due to passive disease-site targeting. In a preclinical liver tumor model,
SNALP-formulated PLK1 siRNA (TKM-080301) demonstrated potent antitumor activity
and induced no measurable immune reaction, minimizing potential nonspecific effects
[36]. A Phase I clinical trial of TKM-080301 was performed in patients with hepatic me-
tastasis from colorectal, pancreas, gastric, breast, and ovarian cancer, but it was termi-
nated without report of results (NCT01437007).
The unique TME of the PDAC make it difficult to deliver therapeutic agents to the
tumor cells and raises the need for the development of a novel therapy. Although the role
of hypoxia-inducible factor 1 (HIF1, including HIF1α and HIF1β) in PDAC development
mechanisms is not completely understood, HIF1α stabilization in the hypoxic TME is
associated with transcriptional activation of multiple signaling pathways involved in the
regulation of cell survival, tumor invasion, angiogenesis, and metabolism [46]. Zhao et al.
developed lipid-polymer hybrid NPs (LENPs), which consist of a single layer or bilayer
lipid shell around a polymeric core made from cationic ε-polylysine co-polymer (ENPs)
[47]. Negatively charged si-HIF1α is absorbed on the surface of ENPs and gemcitabine
encapsulated to the hydrophilic core. LENPs protected NP aggregation, have prolonged
lifetimes with a half-lifetime longer than 3 h, and improved drug release through an en-
hanced tumor vasculature effect within tumor tissues. Treatment with
LENP-Gem-si-HIF1α results in effective silencing of HIF1α both in vitro and in vivo as
well as significant synergistic tumor growth inhibition. Another hypoxia-associated ma-
jor transcription factor, HIF2α (also called endothelial PAS domain protein 1 [EPAS1]) in
pancreatic cancer, was targeted using NPs with siRNA loaded onto a polyethyl-
enimine-poly(lactide-coglycolide) (PLGA)/poloxamer [48]. Treatment inhibited PDAC
cell proliferation by apoptosis induction under hypoxic conditions in vitro, and signifi-
cantly inhibited microvascular formation and tumor growth in orthotopic PDAC mice.
The cytoskeleton, which included microtubules, is composed of α- and β-tubulin
heterodimers. Increased βIII-tubulin expression is associated with poor OS and drug re-
sistance in various types of cancer including pancreatic cancer, and silencing of
βIII-tubulin with shRNA leads to tumor growth inhibition and enhanced sensitivity to
chemotherapy in vitro and in vivo, which suggests the possibility of a novel therapeutic
target for PDAC [49]. The limitations of using highly charged cationic NPs as a delivery
system for siRNA include their toxicity and potential to interact with serum proteins in
the bloodstream. To overcome this problem, Teo et al. developed star polymers with
different lengths of cationic poly(dimethylaminoethyl methacrylate) sidearms and varied
amounts of poly(oligo(ethylene glycol) methyl ether methacrylate), which were highly
accumulated in orthotopic PDAC tumors in mice and had a silencing efficiency of 80% at
the gene and protein levels [50].
3. CRISPR-Cas Gene Editing
CRISPR functions as an immune system in E. coli [51,52]. Recently, the CRISPR systems
have been developed as tools in gene therapy and have shown good target gene specificity,
high editing efficiency, and research simplicity compared to zinc finger nuclease and tran-
scription activator-like effector nuclease [53,54]. CRISPR/Cas9 is an RNA-guided endonucle-
ase consisting of Cas9 protein and sgRNA with target sequence specificity for DNA cleavage.
DNA damage induced by Cas9 is repaired through a non-homologous end-joining DNA
repair pathway or homology-directed repair pathway (HDR), which are cellular DNA repair
mechanisms [55,56]. CRISPR variants have been developed based on this system. Base edit-
ing system (cysteine and adenine base editor) is composed of a single-strand DNA nickase
and deaminase, which can be applied for the treatment of diseases attributable to a point
mutation [5759]. Cas13 (previously referred to as C2C2) edits RNA and reduces the risk for
DNA damage due to off-target effects [60,61]. The process of prime editing involves DNA
Pharmaceutics 2022, 14, 137 7 of 16
nickase and a reverse transcriptase enzyme, which generates new DNA by duplicating an
external RNA template [57,62]. It can lead to the formation of indels (insertions, deletions)
and base conversions without the DNA double-strand breaks or donor DNA templates.
However, it needs an appropriate delivery system for the treatment of cancer. Viral vectors
(retrovirus, adenovirus, adeno-associated virus vector) are popular in the gene therapy field
[6365] but their clinical use is limited by several factors, such as the increased adaptive im-
mune response from neutralizing antibodies by the capsids of adeno-associated virus, the
potential immunogenicity, and gene mutations by their chromosomal insertion [6670].
Therefore, an ideal vehicle would show target specificity, minimize off-target efficiency, and
retain non-immunogenicity.
CRISPR systems can be applied in PDAC gene therapy using three types of nanocarriers:
plasmid DNA (Cas9/sgRNA plasmid), in vitro transcribed Cas9 mRNA/sgRNA, and
pre-assembled ribonucleoprotein (RNP) complex (Figure 1). In general, the successful deliv-
ery of three forms requires cationic coatings to fully condense them [71]. Negatively charged
plasmid DNAs, mRNAs, and RNPs are coated with cationic molecules to form complexes
with structural stability. The complexes bind to cell membranes through electrostatic interac-
tions and are taken into cells via endocytosis. Typically, cationic lipid-based polymers (poly-
ethylenimine, PEI) and peptides (protamine) form positively charged particles that improve
the delivery efficiency and stability in vivo. Cationic lipid-based NPs have additional prop-
erties such as tumor targeting, co-encapsulation of chemotherapeutic agents, and others [72
75]. PEI and protamine are also the most commonly used cationic molecules in vitro and in
vivo. They principally consist of high-density amine groups that interact with negatively
charged nucleic acids or RNPs. They enhance intracellular delivery efficiency and endosomal
escape at a lower pH due to the proton sponge effect [76,77]. In addition, inorganic materials,
such as gold (Au) NPs, are attractive for the delivery of RNPs and donor DNA into cells for
HDR repair. Au NPs are easily modified with thiol-terminated single-stranded DNA and
bind RNPs via non-specific electrostatic forces [78,79].
Figure 1. The cell delivery pathway of CRISPR/Cas9 encapsulated-nanoparticles. The CRISPR sys-
tems can be applied for gene therapy with non-viral carriers in three forms that include: plasmid
DNA (Cas9/sgRNA plasmid), in vitro transcribed Cas9 mRNA/sgRNA, and pre-assembled ribo-
nucleoprotein (RNP) complex. Such nanoparticles can take several forms, including PEGylated- or
cationic lipid-NPs (liposome), cell-mediated exosome, cationic polymers (polyethyleneimine, PEI;
protamine, PA), and cationic lipid- or polymer-coated gold NPs. The plasmid DNA, mRNA and
RNP of NPs may enter the cell via endocytosis and endosome escape, processing the RNP expres-
sion and gene editing.
Pharmaceutics 2022, 14, 137 8 of 16
3.1. Plasmid DNA (Cas9/sgRNA plasmid) Delivery
Although viral vectors such as adenoviruses and lentiviruses are efficient delivery
systems for CRISPR DNA, their immunogenicities make them potential carcinogens and
they have considerable molecular weights [80]. Furthermore, the CRISPR/Cas9 mRNA
and protein delivery remains challenging due to issues related to stability, encapsulation
efficiency, and their high cost of production. CRISPR/Cas9 plasmid delivery has been
used due to the low cost, storage stability, and the potential of prolonged expression.
However, plasmid delivery risks genomic integration, cell stress, and off-target effects
due to long-term transgene expression [81,82].
Plasmid DNA-encoded Cas9 nuclease and sgRNA-encapsulated lipid-based NPs
have been successfully studied in vivo for PDAC treatment. Li et al. reported a tu-
mor-targeted lipid-based CRISPR/Cas9 plasmid delivery system developed to suppress
HIF- (Figure 2a) [71]. They synthesized the R8-dGR peptide (Cys-RRRRRRRRdGR)
bound to integrin αvβ3 and neuropilin-1 in over-expressed tumor cells, and it enhanced
in vivo targeting of pancreatic cancer. The peptide was utilized to lower the charge den-
sity of DOTAP-based cationic liposome. In addition, this cell penetrating peptide en-
hanced cell internalization and transfection efficiency (Figure 2b). HIF-
over-expression in tumors is associated with aberrant p53 and upregulates angiogenesis
and metastasis-related signals such as vascular endothelial growth factor (VEGF) and
matrix metalloproteinase 9 (MMP9) [83,84]. Therefore, the Cas9-HIF- plasmid
DNA/protamine complex and paclitaxel (PTX) co-encapsulated liposome to promote the
anti-metastatic effects could suppress tumor growth in vivo (Figure 2c, d). Consequently,
R8-dGR-Lip/pHIF- and PTX downregulated HIF- and its downstream molecules
VEGF and MMP-9, leading to inhibition of metastasis to livers and lungs (Figure 2e, f).
Figure 2. R8-dGR peptide modified and loaded CRISPR/Cas9-HIF-and paclitaxel cationic lipo-
some for targeted pancreatic treatment. (a) Scheme of the structure of the R8-dGR-Lip/pHIF-
and paclitaxel (PTX) delivery in pancreatic cancer cells. (b) Cellular internalization of with or
without the R8-dGR peptide (Cys-RRRRRRRRdGR) on BxPC-3 cells. scale bar: 100 μm. (c) Tumor
volume of BxPC-3 xenograft models treated with different formulations (mean ± SD, n = 10, *p <
0.05). (d) In vivo downregulation of HIF-by pHIF-1α in tumor tissues. (ef) MMP9 expression
in liver (e) and lung (f) of mice. Scale bar: 100 μm (adapted from Ref. [71]).
Pharmaceutics 2022, 14, 137 9 of 16
Exosomes are naturally released to nano-sized extracellular vesicles from all cells
and contain DNA, RNA, metabolites, cell-surface protein, and lipids depending on the
cell origin [85]. Unlike synthetic nanocarriers, exosomes originating from cells have the
advantage of biocompatibility, non-immunogenicity, and non-cytotoxicity [86]. In addi-
tion, they are contained in various plasma membranes and have enhanced half-live [87].
Therefore, they are suitable as carriers of internal cargo to recipient cells. Recently,
McAndrews et al. reported that exosomes could be successfully engineered to encapsu-
late CRISPR/Cas9 plasmid DNA and delivery it into tumor cells. The exosomes were de-
rived from bone marrow-derived MSCs and did not show repeated dose toxicity [86].
They suppressed the mutant KRASG12D oncogene, leading to inhibited tumor growth in a
syngeneic allograft model and orthotropic model (Figure 3) [88].
Figure 3. Exosome mediated treatment of CRISPR/Cas9 in as allograft and orthotropic model of
pancreatic cancer. (a,b) Tumor volume (a) and tumor weight (b) after exosomes-Cas9/KRASG12D
sgRNA intravenously administration in allograft models (KPC689 cells) every other day for 2
weeks (n = 8 mice in each group). (c) Cas9 mRNA expression levels in tumor tissues by quantita-
tive PCR (normalized to 18S, n = 5 mice per group). (d) Tumor growth by bioluminescent imaging
after exsomes-Cas9/KRASG12D sgRNA Intraperitoneal injection in orthotopic models
(KPC689-GFP-Luciferase cells) every other day for 3 weeks. (e) KRASG12D mRNA expression
levels at the end point in orthotropic tumor tissues (mean ± SD, *p < 0.05, **p < 0.01) (adapted from
Ref. [88]).
3.2. In Vitro Transcribed Cas9/sgRNA mRNA Delivery
CRISPR/Cas9 mRNA delivery has advantages over plasmid DNA. The mRNA is not
integrated into the target genome, which prevents off-target effects [65,89,90]. In addi-
tion, the mRNA leads to quick and transient Cas9 protein expression in the cytoplasm.
However, mRNAs are large in size, unstable, and susceptible to degradation during de-
livery [91].
Pharmaceutics 2022, 14, 137 10 of 16
3.3. Pre-Assembled Ribonucleoprotein (RNP) Complex Delivery
CRISPR/Cas9 RNP does not need to be translated into cells. It is directly transported
into the nucleus and edits the target gene. In addition, it has low toxicity, high gene-editing
efficiency, low off-target effects, and no chance of integration into the genome of recipient
cells [65,89,92,93]. The Cas9 protein and sgRNA can be simply mixed ex vivo for delivery to
cells. The Cas9 protein/sgRNA complex is negatively charged. The complex can be positively
charged by modification to facilitate cell membrane affinity and internalization [92,9497]. In
addition, bio-reducible disulfide bonds of nanocarriers can be adjusted to improve degrada-
tion and endosomal escape [98]. In addition, the Cas9 protein can be modified to enhance the
encapsulation efficiency of NPs [99,100].
Zhao et al. studied CRISPR-Cas13a effector protein delivered to pancreatic cancer
cells using lipofectamine CRISPRMAX and found that the Cas13a-crRNA complex bound
to and cleaved target RNA and xenograft model by intratumoral injection (Figure 4a)
[101]. CRISPR-Cas13a system reduces mRNA expression in mammalian cells and shows
an improved efficacy and specificity over RNA interference [60,61]. The system reduced
the KRASG12D mRNA expression with up to 70% knockdown efficiency (Fig, 4bd), lead-
ing to apoptosis and tumor growth inhibition (Figure 4e, f) [101].
Figure 4. CRISPR/Cas13a-lipofectamine CRISPRMAX delivery for KRAS mutated pancreatic cancer
treatment. (a) The scheme of Cas13 system differentiates wild type (WT, normal cells) and mutated
KRAS genes (cancer cells). (b) crRNA sequences of the crRNA19-target sequence in KRAS-G12D and
KRAS WT. (c) qRT-PCR analysis of KRAS-G12D mRNA expression after crRNA19-14 delivery into
KRAS-G12D mutated cells. (d) qRT-PCR analysis of KRAS-WT mRNA expression after crRNA19-14
delivery into KRAS-WT cells. (e) The tumor volumes of subcutaneous AsPC-1 xenograft after admin-
istration with repeated intratumoral injections of the Cas13a-lipofectamine for 21 days. (f) Staining for
apoptotic cells in tumor tissues (green fluorescence). Scale bar : 50 μm. (mean ± SD, *p < 0.05, **p < 0.01,
and ***p < 0.001) (adapted from Ref [101]).
Recently, our group has developed a tumor targeted-nanoliposome
(NL[Cas9/ABE]-Ab) that simultaneously encapsulates CRISPR/Cas9 and adenine-base editor
(ABE) proteins for dual-gene editing (Figure 5) [15]. The Cas9 and ABE proteins are modified
with his-tag protein to improve NL-encapsulation efficiency. In addition, we used
PEG-bound lipid for structure stability and increased receptor-mediated endocytosis. Gem-
citabine is used as first-line chemotherapy for PDAC but is limited in that most PDAC pa-
tients develop resistance to it [102]. The KRAS mutation and loss of P53 induces HIF-1α sta-
Pharmaceutics 2022, 14, 137 11 of 16
bilization, which is the master regulator of glucose metabolism and a major driver of gem-
citabine chemoresistance in PDAC [103,104]. Co-administration of NL(Cas9/ABE)-Ab and
gemcitabine markedly inhibits tumor proliferation in a PANC1 xenograft (Figure 5c).
NL(Cas9/ABE)-Ab inhibits KRAS and TP53 mutations, and regulates HIF--related glycol-
ysis, which promotes gemcitabine sensitivity in vivo (Figure 5d-f). Therefore, the dual-gene
editing tool of the KRAS mutation and mutant TP53 might overcome drug-resistance in
PDAC [15].
Figure 5. Dual-gene editing nanoliposome system as an overcoming drug-resistance for PDAC. (a)
Graphical summary of NL(Cas9/ABE)-Ab composition. (b) Dynamic distributions of tumor targeting
efficiency by NL with or without Ab in vivo. Mice were injected intraperitoneally once with NL-Ab la-
beled with RITC. PANC1 xenograft tumor was marked as white circles. (c) The scheme of PANC1
xenografts co-administrated with NL(Cas9/ABE)-Ab and gemcitabine. The NL(Cas9/ABE)-Ab was in-
jected intraperitoneally once (red arrow) and then gemcitabine (50 mg/kg) was administered twice a
week for 4 weeks. Mean ± SEM, n = 3 per group, *p < 0.05, ***p < 0.001. (d) Immunofluorescence with
mutant P53 and KRAS on the tumor sections. Scale bar: 50 μm. (e) Immunostaining with HIF1a-related
glucose metabolism and gemcitabine transporter (GLUT1, TKT, CTPS and ENT1) on the tumor. Scale
bar: 50 μm. (f) Immunostaining with apoptotic cells (TUNEL assay and Ki67) by NL(Cas9/ABE)-Ab
and gemcitabine on the tumor. Scale bar: 100 μm (adapted from Ref. [15]).
Pharmaceutics 2022, 14, 137 12 of 16
4. Conclusions and Future Perspectives
Prerequisites for successful gene therapy include rapid, simple, and easy synthesis,
safety, and efficient delivery. Gene therapy using RNAi-based therapeutics and CRISPR
genome editing is promising and can improve precise gene delivery to tumor tissue.
Although there have been major advances in the development of gene therapy for the
PDAC treatment, there is still much room for improvement in pharmacokinetics, phar-
macodynamics, and methods to improve safety. RNAi-based therapeutics have several
barriers (e.g., systemic circulation and targeted delivery must be improved, off-target
effects must be minimized, etc.) and it is essential to further develop safe, biocompatible,
and biodegradable NP-based therapeutics for clinical application. EPR of nanoparti-
cle-based carriers make it easier to accumulate in the tumor cells. However, extracellular
and intracellular barriers in PDAC are hurdles to overcome. Nanoparticles with appro-
priate particle size and their chemical modifications and functionalization with targeting
ligands are necessary to enhance the uptakes into tumor cells and address other biologi-
cal issues such as intracellular trafficking and intracellular siRNA escape [105]. In addi-
tion, combination treatment with chemotherapy and RNAi-based therapeutics or a
strategy combining RNAi molecules and anticancer drugs would help overcome drug
resistance and TME barriers.
CRISPR/Cas9 genome editing has widely been used not only in the development of
cancer models and as a tool for the identification and validation of therapeutic targets but
also as a potential cancer therapeutic [106]. The field of cancer immunotherapy is the
most advanced clinical application. The first Phase I trial from China including 22 pa-
tients with NSLC used CRISPR-Cas9 PD-1-edited T cells from patient blood, and the
treatments were safe and showed some therapeutic efficacy (NCT02793856) [107]. Sever-
al clinical trials are ongoing to demonstrate the efficacy of more potent chimeric antigen
receptor T cells using CRISPR to knock out immune co-inhibitory pathways or signaling
molecules [108]. However, there are several huddles to cross before CRISPR/Cas9 ge-
nome editing can be used in clinical practice, including potential off-target effects and
safety issues. A novel therapeutic approach is required for efficient delivery, accurate
targeting of desired cells, efficient gene editing, and to minimize off-target effects and
immune responses. Although several clinical trials for gene therapy have not been suc-
cessful, the progress in understanding of the PDAC TME and nanomedicine-based gene
therapy can make it possible to improve the clinical outcomes of patients with PDAC.
Author Contributions: Conceptualization, E.-J.W. and Y.-S.C.; writingoriginal draft preparation,
E.-J.W., H.P., and Y.-S.C.; writingreview and editing, E.-J.W., H.P., T.-J.Y., and Y.-S.C.; supervi-
sion, T.-J.Y. and Y.-S.C. All authors discussed, edited, and approved the final version of the man-
uscript. All authors have read and agreed to the published version of the manuscript.
Funding: This study was supported by a grant of the National Research Foundation of Korea
(grant number 2019R1F1A1058879).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: The data presented in this study are available in this manuscript.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Park, W.; Chawla, A.; O’Reilly, E.M. Pancreatic Cancer: A Review. JAMA 2021, 326, 851862.
2. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 20082020.
3. Strobel, O.; Neoptolemos, J.; Jäger, D.; Büchler, M.W. Optimizing the outcomes of pancreatic cancer surgery. Nat. Rev. Clin.
Oncol. 2019, 16, 1126.
4. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et
al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 23952406.
Pharmaceutics 2022, 14, 137 13 of 16
5. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.L.; Gourgou-Bourgade, S.;
de la Fouchardière, C.; et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364,
18171825.
6. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.;
et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 2013, 369, 16911703.
7. Neoptolemos, J.P.; Kleeff, J.; Michl, P.; Costello, E.; Greenhalf, W.; Palmer, D.H. Therapeutic developments in pancreatic can-
cer: Current and future perspectives. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 333348.
8. Pook, H.; Pauklin, S. Mechanisms of Cancer Cell Death: Therapeutic Implications for Pancreatic Ductal Adenocarcinoma.
Cancers 2021, 13, 4834.
9. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 2014, 371, 10391049.
10. Hosein, A.N.; Brekken, R.A.; Maitra, A. Pancreatic cancer stroma: An update on therapeutic targeting strategies. Nat. Rev.
Gastroenterol. Hepatol. 2020, 17, 487505.
11. Cannon, A.; Thompson, C.; Hall, B.R.; Jain, M.; Kumar, S.; Batra, S.K. Desmoplasia in pancreatic ductal adenocarcinoma: In-
sight into pathological function and therapeutic potential. Genes Cancer 2018, 9, 7886.
12. Nevala-Plagemann, C.; Hidalgo, M.; Garrido-Laguna, I. From state-of-the-art treatments to novel therapies for advanced-stage
pancreatic cancer. Nat. Rev. Clin. Oncol. 2020, 17, 108123.
13. El-Zahaby, S.A.; Elnaggar, Y.S.R.; Abdallah, O.Y. Reviewing two decades of nanomedicine implementations in targeted
treatment and diagnosis of pancreatic cancer: An emphasis on state of art. J. Control. Release 2019, 293, 2135.
14. Hu, X.; Xia, F.; Lee, J.; Li, F.; Lu, X.; Zhuo, X.; Nie, G.; Ling, D. Tailor-Made Nanomaterials for Diagnosis and Therapy of Pan-
creatic Ductal Adenocarcinoma. Adv. Sci. 2021, 8, 2002545.
15. Won, E.J.; Park, H.; Chang, S.H.; Kim, J.H.; Kwon, H.; Cho, Y.S.; Yoon, T.J. One-shot dual gene editing for drug-resistant pan-
creatic cancer therapy. Biomaterials 2021, 279, 121252.
16. Hannon, G.J. RNA interference. Nature 2002, 418, 244251.
17. Adams, D.; Gonzalez-Duarte, A.; O’Riordan, W.D.; Yang, C.C.; Ueda, M.; Kristen, A.V.; Tournev, I.; Schmidt, H.H.; Coelho, T.;
Berk, J.L.; et al. Patisiran, an RNAi Therapeutic, for Hereditary Transthyretin Amyloidosis. N. Engl. J. Med. 2018, 379, 1121.
18. Hoy, S.M. Patisiran: First, Global Approval. Drugs 2018, 78, 16251631.
19. Goodall, G.J.; Wickramasinghe, V.O. RNA in cancer. Nat. Rev. Cancer 2021, 21, 2236.
20. Hernandez, Y.G.; Lucas, A.L. MicroRNA in pancreatic ductal adenocarcinoma and its precursor lesions. World J. Gastrointest.
Oncol. 2016, 8, 1829.
21. Gilles, M.E.; Hao, L.; Huang, L.; Rupaimoole, R.; Lopez-Casas, P.P.; Pulver, E.; Jeong, J.C.; Muthuswamy, S.K.; Hidalgo, M.;
Bhatia, S.N.; et al. Personalized RNA Medicine for Pancreatic Cancer. Clin. Cancer Res. 2018, 24, 17341747.
22. Goswami, C.P.; Nakshatri, H. PROGmiR: A tool for identifying prognostic miRNA biomarkers in multiple cancers using pub-
licly available data. J. Clin. Bioinform. 2012, 2, 23.
23. Li, Y.; Chen, Y.; Li, J.; Zhang, Z.; Huang, C.; Lian, G.; Yang, K.; Chen, S.; Lin, Y.; Wang, L.; et al. Co-delivery of microRNA-21
antisense oligonucleotides and gemcitabine using nanomedicine for pancreatic cancer therapy. Cancer Sci. 2017, 108, 1493
1503.
24. Xie, Y.; Hang, Y.; Wang, Y.; Sleightholm, R.; Prajapati, D.R.; Bader, J.; Yu, A.; Tang, W.; Jaramillo, L.; Li, J.; et al. Stromal Mod-
ulation and Treatment of Metastatic Pancreatic Cancer with Local Intraperitoneal Triple miRNA/siRNA Nanotherapy. ACS
Nano 2020, 14, 255271.
25. Wu, Y.; Tang, Y.; Xie, S.; Zheng, X.; Zhang, S.; Mao, J.; Wang, B.; Hou, Y.; Hu, L.; Chai, K.; et al. Chimeric peptide supramolec-
ular nanoparticles for plectin-1 targeted miRNA-9 delivery in pancreatic cancer. Theranostics 2020, 10, 11511165.
26. Srivastava, S.K.; Bhardwaj, A.; Singh, S.; Arora, S.; Wang, B.; Grizzle, W.E.; Singh, A.P. MicroRNA-150 directly targets MUC4
and suppresses growth and malignant behavior of pancreatic cancer cells. Carcinogenesis 2011, 32, 18321839.
27. Arora, S.; Swaminathan, S.K.; Kirtane, A.; Srivastava, S.K.; Bhardwaj, A.; Singh, S.; Panyam, J.; Singh, A.P. Synthesis, char-
acterization, and evaluation of poly (D,L-lactide-co-glycolide)-based nanoformulation of miRNA-150: Potential implications
for pancreatic cancer therapy. Int. J. Nanomed. 2014, 9, 29332942.
28. Zhang, S.; Cheng, Z.; Wang, Y.; Han, T. The Risks of miRNA Therapeutics: In a Drug Target Perspective. Drug. Des. Dev. Ther.
2021, 15, 721733.
29. Setten, R.L.; Rossi, J.J.; Han, S.P. The current state and future directions of RNAi-based therapeutics. Nat. Rev. Drug. Discov.
2019, 18, 421446.
30. Nakamura, Y.; Mochida, A.; Choyke, P.L.; Kobayashi, H. Nanodrug Delivery: Is the Enhanced Permeability and Retention
Effect Sufficient for Curing Cancer? Bioconjugate Chem. 2016, 27, 22252238.
31. Hattab, D.; Gazzali, A.M.; Bakhtiar, A. Clinical Advances of siRNA-Based Nanotherapeutics for Cancer Treatment. Pharma-
ceutics 2021, 13, 1009.
32. Zuckerman, J.E.; Gritli, I.; Tolcher, A.; Heidel, J.D.; Lim, D.; Morgan, R.; Chmielowski, B.; Ribas, A.; Davis, M.E.; Yen, Y. Cor-
relating animal and human phase Ia/Ib clinical data with CALAA-01, a targeted, polymer-based nanoparticle containing
siRNA. Proc. Natl. Acad. Sci. USA 2014, 111, 1144911454.
33. Schultheis, B.; Strumberg, D.; Santel, A.; Vank, C.; Gebhardt, F.; Keil, O.; Lange, C.; Giese, K.; Kaufmann, J.; Khan, M.; et al.
First-in-human phase I study of the liposomal RNA interference therapeutic Atu027 in patients with advanced solid tumors. J.
Clin. Oncol. 2014, 32, 41414148.
Pharmaceutics 2022, 14, 137 14 of 16
34. Schultheis, B.; Strumberg, D.; Kuhlmann, J.; Wolf, M.; Link, K.; Seufferlein, T.; Kaufmann, J.; Feist, M.; Gebhardt, F.; Khan, M.;
et al. Safety, Efficacy and Pharcacokinetics of Targeted Therapy with The Liposomal RNA Interference Therapeutic Atu027
Combined with Gemcitabine in Patients with Pancreatic Adenocarcinoma. A Randomized Phase Ib/IIa Study. Cancers 2020,
12, 3130.
35. Golan, T.; Khvalevsky, E.Z.; Hubert, A.; Gabai, R.M.; Hen, N.; Segal, A.; Domb, A.; Harari, G.; David, E.B.; Raskin, S.; et a l.
RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget
2015, 6, 2456024570.
36. Judge, A.D.; Robbins, M.; Tavakoli, I.; Levi, J.; Hu, L.; Fronda, A.; Ambegia, E.; McClintock, K.; MacLachlan, I. Confirming the
RNAi-mediated mechanism of action of siRNA-based cancer therapeutics in mice. J. Clin. Investig. 2009, 119, 661673.
37. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate thera peutic
targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498503.
38. O’Brien, Z.; Wang, L.; Majeti, B.; Clamme, J.; Baclig, R.; Chu, J.; Fong, S.; Harborth, J.; Ibarra, J.; Yin, H.; et al. A novel lipid
nanoparticle (NBF-006) encapsulating glutathione Stransferase P (GSTP) siRNA for the treatment of KRAS-driven non-small
cell lung cancer. Cancer Res. 2018, 78, 5917.
39. Chatterjee, A.; Gupta, S. The multifaceted role of glutathione S-transferases in cancer. Cancer Lett. 2018, 433, 3342.
40. Khvalevsky, E.Z.; Gabai, R.; Rachmut, I.H.; Horwitz, E.; Brunschwig, Z.; Orbach, A.; Shemi, A.; Golan, T.; Domb, A.J.; Yavin,
E.; et al. Mutant KRAS is a druggable target for pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 2072320728.
41. Aye, Y.; Li, M.; Long, M.J.; Weiss, R.S. Ribonucleotide reductase and cancer: Biological mechanisms and targeted therapies.
Oncogene 2015, 34, 20112021.
42. Davis, M.E.; Zuckerman, J.E.; Choi, C.H.J.; Seligson, D.; Tolcher, A.; Alabi, C.A.; Yen, Y.; Heidel, J.D.; Ribas, A. Evidence of
RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature 2010, 464, 10671070.
43. Aleku, M.; Schulz, P.; Keil, O.; Santel, A.; Schaeper, U.; Dieckhoff, B.; Janke, O.; Endruschat, J.; Durieux, B.; Röder, N; e t al.
Atu027, a liposomal small interfering RNA formulation targeting protein kinase N3, inhibits cancer progression. Cancer Res.
2008, 68, 97889798.
44. Santel, A.; Aleku, M.; Röder, N.; Möpert, K.; Durieux, B.; Janke, O.; Keil, O.; Endruschat, J.; Dames, S.; Lange, C; et al. Atu027
prevents pulmonary metastasis in experimental and spontaneous mouse metastasis models. Clin. Cancer. Res. 2010, 16, 5469
5480.
45. Strebhardt, K.; Ullrich, A. Targeting polo-like kinase 1 for cancer therapy. Nat. Rev. Cancer 2006, 6, 321330.
46. Shukla, S.K.; Purohit, V.; Mehla, K.; Gunda, V.; Chaika, N.V.; Vernucci, E.; King, R.J.; Abrego, J.; Goode, G.D.; Dasgupta, A.; et
al. MUC1 and HIF-1alpha Signaling Crosstalk Induces Anabolic Glucose Metabolism to Impart Gemcitabine Resistance to
Pancreatic Cancer. Cancer Cell 2017, 32, 392.
47. Zhao, X.; Li, F.; Li, Y.; Wang, H.; Ren, H.; Chen, J.; Nie, G.; Hao, J. Co-delivery of HIF1α siRNA and gemcitabine via biocom-
patible lipid-polymer hybrid nanoparticles for effective treatment of pancreatic cancer. Biomaterials 2015, 46, 1325.
48. Pan, X.; Zhu, Q.; Sun, Y.; Li, L.; Zhu, Y.; Zhao, Z.; Zuo, J.; Fang, W.; Li, K. PLGA/poloxamer nanoparticles loaded with EPAS1
siRNA for the treatment of pancreatic cancer in vitro and in vivo. Int. J. Mol. Med. 2015, 35, 9951002.
49. McCarroll, J.A.; Sharbeen, G.; Liu, J.; Youkhana, J.; Goldstein, D.; McCarthy, N.; Limbri, L.F.; Dischl, D.; Ceyhan, G.O.; Erkan,
M.; et al. βIII-tubulin: A novel mediator of chemoresistance and metastases in pancreatic cancer. Oncotarget 2015, 6, 22352249.
50. Teo, J.; McCarroll, J.A.; Boyer, C.; Youkhana, J.; Sagnella, S.M.; Duong, H.T.; Liu, J.; Sharbeen, G.; Goldstein, D.; Davis, T.P.; et
al. A Rationally Optimized Nanoparticle System for the Delivery of RNA Interference Therapeutics into Pancreatic Tumors In
Vivo. Biomacromolecules 2016, 17, 23372351.
51. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA en-
donuclease in adaptive bacterial immunity. Science 2012, 337, 816821.
52. Ran, F.A.; Hsu, P.D.; Wright, J.; Agarwala, V.; Scott, D.A.; Zhang, F. Genome engineering using the CRISPR-Cas9 system. Nat.
Protoc. 2013, 8, 22812308.
53. Gaj, T.; Gersbach, C.A.; Barbas, C.F.; III. ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends Bio-
technol. 2013, 31, 397405.
54. Osborn, M.J.; Belanto, J.J.; Tolar, J.; Voytas, D.F. Gene editing and its application for hematological diseases. Int. J. Hematol.
2016, 104, 1828.
55. Jiang, F.; Doudna, J.A. CRISPR-Cas9 Structures and Mechanisms. Annu. Rev. Biophys. 2017, 46, 505529.
56. Kaboli, S.; Babazada, H. CRISPR Mediated Genome Engineering and its Application in Industry. Curr. Issues Mol. Biol. 2018,
26, 8192.
57. Anzalone, A.V.; Koblan, L.W.; Liu, D.R. Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime
editors. Nat. Biotechnol. 2020, 38, 824844.
58. Gaudelli, N.M.; Komor, A.C.; Rees, H.A.; Packer, M.S.; Badran, A.H.; Bryson, D.I.; Liu, D.R. Programmable base editing of
A•T to G•C in genomic DNA without DNA cleavage. Nature 2017, 551, 464471.
59. Komor, A.C.; Kim, Y.B.; Packer, M.S.; Zuris, J.A.; Liu, D.R. Programmable editing of a target base in genomic DNA without
double-stranded DNA cleavage. Nature 2016, 533, 420424.
60. Abudayyeh, O.O.; Gootenberg, J.S.; Essletzbichler, P.; Han, S.; Joung, J.; Belanto, J.J.; Verdine, V.; Cox, D.B.; Kellner, M.J.; Re-
gev, A.; et al. RNA targeting with CRISPR-Cas13. Nature 2017, 550, 280284.
Pharmaceutics 2022, 14, 137 15 of 16
61. Cox, D.B.; Gootenberg, J.S.; Abudayyeh, O.O.; Franklin, B.; Kellner, M.J.; Joung, J.; Zhang, F. RNA editing with CRISPR-Cas13.
Science 2017, 358, 10191027.
62. Anzalone, A.V.; Randolph, P.B.; Davis, J.R.; Sousa, A.A.; Koblan, L.W.; Levy, J.M.; Chen, P.J.; Wilson, C.; Newby, G.A.; Ragu-
ram, A.; et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019, 576, 149157.
63. Nelson, C.E.; Gersbach, C.A. Engineering Delivery Vehicles for Genome Editing. Annu. Rev. Chem. Biomol. Eng. 2016, 7, 637662.
64. Yin, H.; Kauffman, K.J.; Anderson, D.G. Delivery technologies for genome editing. Nat. Rev. Drug Discov. 2017, 16, 387399.
65. Li, L.; Hu, S.; Chen, X. Non-viral delivery systems for CRISPR/Cas9-based genome editing: Challenges and opportunities.
Biomaterials 2018, 171, 207218.
66. Louis Jeune, V.; Joergensen, J.A.; Hajjar, R.J.; Weber, T. Pre-existing anti-adeno-associated virus antibodies as a challenge in
AAV gene therapy. Hum. Gene Ther. Methods 2013, 24, 5967.
67. Rapti, K.; Louis-Jeune, V.; Kohlbrenner, E.; Ishikawa, K.; Ladage, D.; Zolotukhin, S.; Hajjar, R.J.; Weber, T. Neutralizing anti-
bodies against AAV serotypes 1, 2, 6, and 9 in sera of commonly used animal models. Mol. Ther. 2012, 20, 7383.
68. Li, L.; He, Z.Y.; Wei, X.W.; Gao, G.P.; Wei,Y.Q. Challenges in CRISPR/CAS9 Delivery: Potential Roles of Nonviral Vectors.
Hum. Gene Ther. 2015, 26, 452462.
69. Wang, D.; Mou, H.; Li, S.; Li, Y.; Hough, S.; Tran, K.; Li, J.; Yin, H.; Anderson, D.G.; Sontheimer, E.J.; et al. Adenovi-
rus-Mediated Somatic Genome Editing of Pten by CRISPR/Cas9 in Mouse Liver in Spite of Cas9-Specific Immune Responses.
Hum. Gene Ther. 2015, 26, 432442.
70. Lao, Y.H.; Li, M.; Gao, M.A.; Shao, D.; Chi, C.W.; Huang, D.; Chakraborty, S.; Ho, T.C.; Jiang, W.; Wang, H.X.; et al. HPV On-
cogene Manipulation Using Nonvirally Delivered CRISPR/Cas9 or Natronobacterium gregoryi Argonaute. Adv. Sci. 2018, 5,
1700540.
71. Li, M.; Xie, H.; Liu, Y.; Xia, C.; Cun, X.; Long, Y.; Chen, X.; Deng, M.; Guo, R.; Zhang, Z.; et al. Knockdown of hypox-
ia-inducible factor-1 alpha by tumor targeted delivery of CRISPR/Cas9 system suppressed the metastasis of pancreatic cancer.
J. Control. Release 2019, 304, 204215.
72. Luo, Y.L.; Xu, C.F.; Li, H.J.; Cao, Z.T.; Liu, J.; Wang, J.L.; Du, X.J.; Yang, X.Z.; Gu, Z.; et al. Macrophage-Specific in Vivo Gene
Editing Using Cationic Lipid-Assisted Polymeric Nanoparticles. ACS Nano 2018, 12, 9941005.
73. Wang, P.; Zhang, L.; Zheng, W.; Cong, L.; Guo, Z.; Xie, Y.; Wang, L.; Tang, R.; Feng, Q.; Hamada, Y.; et al. Thermo-triggered
Release of CRISPR-Cas9 System by Lipid-Encapsulated Gold Nanoparticles for Tumor Therapy. Angew. Chem. Int. Ed. Engl.
2018, 57, 14911496.
74. Schuh, R.S.; de Carvalho, T.G.; Giugliani, R.; Matte, U.; Baldo, G.; Teixeira, H.F. Gene editing of MPS I human fibroblasts by
co-delivery of a CRISPR/Cas9 plasmid and a donor oligonucleotide using nanoemulsions as nonviral carriers. Eur. J. Pharm.
Biopharm. 2018, 122, 158166.
75. Wang, H.X.; Song, Z.; Lao, Y.H.; Xu, X.; Gong, J.; Cheng, D.; Chakraborty, S.; Park, J.S.; Li, M.; Huang, D.; et al. Nonviral gene
editing via CRISPR/Cas9 delivery by membrane-disruptive and endosomolytic helical polypeptide. Proc. Natl. Acad. Sci. USA
2018, 115, 49034908.
76. Ryu, N.; Kim, M.A.; Park, D.; Lee, B.; Kim, Y.R.; Kim, K.H.; Baek, J.I.; Kim, W.J.; Lee, K.Y.; Kim, U.K. Effective PEI-mediated
delivery of CRISPR-Cas9 complex for targeted gene therapy. Nanomedicine 2018, 14, 20952102.
77. Zhang, L.; Wang, P.; Feng, Q.; Wang, N.; Chen, Z.; Huang, Y.; Zheng, W.; Jiang, X. Lipid nanoparticle-mediated efficient de-
livery of CRISPR/Cas9 for tumor therapy. NPG Asia Mater. 2017, 9, e441.
78. Ding, Y.; Jiang, Z.; Saha, K.; Kim, C.S.; Kim, S.T.; Landis, R.F.; Rotello, V.M. Gold nanoparticles for nucleic acid delivery. Mol.
Ther. 2014, 22, 10751083.
79. Lee, K.; Conboy, M.; Park, H.M.; Jiang, F.; Kim, H.J.; Dewitt, M.A.; Mackley, V.A.; Chang, K.; Rao, A.; Skinner, C.; et al. Na-
noparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nat. Biomed.
Eng. 2017, 1, 889901.
80. Zuckermann, M.; Hovestadt, V.; Knobbe-Thomsen, C.B.; Zapatka, M.; Northcott, P.A.; Schramm, K.; Belic, J.; Jones, D.T.;
Tschida, B.; Moriarity, B.; et al. Somatic CRISPR/Cas9-mediated tumour suppressor disruption enables versatile brain tumour
modelling. Nat. Commun. 2015, 6, 7391.
81. Wang, H.X.; Li, M.; Lee, C.M.; Chakraborty, S.; Kim, H.W.; Bao, G.; Leong, K.W. CRISPR/Cas9-Based Genome Editing for
Disease Modeling and Therapy: Challenges and Opportunities for Nonviral Delivery. Chem. Rev. 2017, 117, 98749906.
82. Ibraheem, D.; Elaissari, A.; Fessi, H. Gene therapy and DNA delivery systems. Int. J. Pharm. 2014, 459, 7083.
83. Guan, G.; Zhang, Y.; Lu, Y.; Liu, L.; Shi, D.; Wen, Y.; Yang, L.; Ma, Q.; Liu, T.; Zhu, X.; et al. The HIF-1α/CXCR4 pathway
supports hypoxia-induced metastasis of human osteosarcoma cells. Cancer Lett. 2015, 357, 254264.
84. Chang, Y.C.; Chan, Y.C.; Chang, W.M.; Lin, Y.F.; Yang, C.J.; Su, C.Y.; Huang, M.S.; Wu, A.T.; Hsiao, M. Feedback regulation of
ALDOA activates the HIF-1α/MMP9 axis to promote lung cancer progression. Cancer Lett. 2017, 403, 2836.
85. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, 6478.
86. Mendt, M.; Kamerkar, S.; Sugimoto, H.; McAndrews, K.M.; Wu, C.C.; Gagea, M.; Yang, S.; Blanko, E.V.R.; Peng, Q.; Ma, X.; et
al. Generation and testing of clinical-grade exosomes for pancreatic cancer. JCI Insight 2018, 3, e99263.
87. Liu, H.; Qiao, S.; Fan, X.; Gu, Y.; Zhang, Y.; Huang, S. Role of exosomes in pancreatic cancer. Oncol. Lett. 2021, 21, 298.
88. McAndrews, K.M.; Xiao, F.; Chronopoulos, A.; LeBleu, V.S.; Kugeratski, F.G.; Kalluri, R. Exosome-mediated delivery of
CRISPR/Cas9 for targeting of oncogenic Kras(G12D) in pancreatic cancer. Life Sci. Alliance 2021, 4.
Pharmaceutics 2022, 14, 137 16 of 16
89. Glass, Z.; Lee, M.; Li, Y.; Xu, Q. Engineering the Delivery System for CRISPR-Based Genome Editing. Trends Biotechnol. 2018,
36, 173185.
90. Liang, X.; Potter, J.; Kumar, S.; Zou, Y.; Quintanilla, R.; Sridharan, M.; Carte, J.; Chen, W.; Roark, N.; Ranganathan, S.; et al.
Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J. Biotechnol. 2015, 208, 4453.
91. Yin, H.; Kanasty, R.L.; Eltoukhy, A.A.; Vegas, A.J.; Dorkin, J.R.; Anderson, D.G. Non-viral vectors for gene-based therapy.
Nat. Rev. Genet. 2014, 15, 541555.
92. Kim, S.; Kim, D.; Cho, S.W.; Kim, J.; Kim, J.S. Highly efficient RNA-guided genome editing in human cells via delivery of pu-
rified Cas9 ribonucleoproteins. Genome Res. 2014, 24, 10121019.
93. Hendel, A.; Bak, R.O.; Clark, J.T.; Kennedy, A.B.; Ryan, D.E.; Roy, S.; Steinfeld, I.; Lunstad, B.D.; Kaiser, R.J.; Wilkens, A.B.; et
al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat. Biotechnol. 2015, 33,
985989.
94. Zhang, S.; Shen, J.; Li, D.; Cheng, Y. Strategies in the delivery of Cas9 ribonucleoprotein for CRISPR/Cas9 genome editing.
Theranostics 2021, 11, 614648.
95. Ramakrishna, S.; Kwaku Dad, A.B.; Beloor, J.; Gopalappa, R.; Lee, S.K.; Kim, H. Gene disruption by cell-penetrating pep-
tide-mediated delivery of Cas9 protein and guide RNA. Genome Res. 2014, 24, 10201027.
96. Zuris, J.A.; Thompson, D.B.; Shu, Y.; Guilinger, J.P.; Bessen, J.L.; Hu, J.H.; Maeder, M.L.; Joung, J.K.; Chen, Z.Y.; Liu, D.R. Cat-
ionic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol.
2015, 33, 7380.
97. Yu, X.; Liang, X.; Xie, H.; Kumar, S.; Ravinder, N.; Potter, J.; du Jeu, X.D.M.; Chesnut, J.D. Improved delivery of Cas9 pro-
tein/gRNA complexes using lipofectamine CRISPRMAX. Biotechnol. Lett. 2016, 38, 919929.
98. Wang, M.; Zuris, J.A.; Meng, F.; Rees, H.; Sun, S.; Deng, P.; Han, Y.; Gao, X.; Pouli, D.; Wu, Q.; et al. Efficient delivery of ge-
nome-editing proteins using bioreducible lipid nanoparticles. Proc. Natl. Acad. Sci. USA 2016, 113, 28682873.
99. Cho, E.Y.; Ryu, J.Y.; Lee, H.A.R.; Hong, S.H.; Park, H.S.; Hong, K.S.; Park, S.G.; Kim, H.P.; Yoon, T.J. Lecithin nano-liposomal
particle as a CRISPR/Cas9 complex delivery system for treating type 2 diabetes. J. Nanobiotechnology 2019, 17, 19.
100. Ryu, J.Y.; Choi, Y.J.; Won, E.J.; Hui, E.; Kim, H.S.; Cho, Y.S.; Yoon, T.J. Gene editing particle system as a therapeutic approach
for drug-resistant colorectal cancer. Nano Res. 2020, 13, 15761585.
101. Zhao, X.; Liu, L.; Lang, J.; Cheng, K.; Wang, Y.; Li, X.; Shi, J.; Wang, Y.; Nie, G. A CRISPR-Cas13a system for efficient and spe-
cific therapeutic targeting of mutant KRAS for pancreatic cancer treatment. Cancer Lett. 2018, 431, 171181.
102. Binenbaum, Y.; Na’ara, S.; Gil, Z. Gemcitabine resistance in pancreatic ductal adenocarcinoma. Drug Resist. Updates 2015, 23,
5568.
103. Yokoi, K.; Fidler, I.J. Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin.
Cancer Res. 2004, 10, 22992306.
104. Poon, E.; Harris, A.L.; Ashcroft, M. Targeting the hypoxia-inducible factor (HIF) pathway in cancer. Expert Rev. Mol. Med.
2009, 11, e26.
105. Aghamiri, S.; Raee, P.; Talaei, S.; MohammadiYeganeh, S.; Bayat, S.; Rezaee, D.; Ghavidel, A.A.; Teymouri, A.; Roshanzamiri,
S.; Farhadi, S.; et al. Nonviral siRNA delivery systems for pancreatic cancer therapy. Biotechnol. Bioeng. 2021, 118, 36693690.
106. Yin, H.; Xue, W.; Anderson, D.G. CRISPR-Cas: A tool for cancer research and therapeutics. Nat. Rev. Clin. Oncol. 2019, 16, 281
295.
107. Lu, Y.; Xue, J.; Deng, T.; Zhou, X.; Yu, K.; Deng, L.; Huang, M.; Yi, X.; Liang, M.; Wang, Y.; et al. Safety and feasibility of
CRISPR-edited T cells in patients with refractory non-small-cell lung cancer. Nat. Med. 2020, 26, 732740.
108. Zhang, M.; Eshraghian, E.A.; Al Jammal, O.; Zhang, Z.; Zhu, X. CRISPR technology: The engine that drives cancer therapy.
Biomed. Pharmacother. 2021, 133, 111007.
... patients with pancreatic ductal adenocarcinoma (PDAC) (27). Furthermore, CD73 deletion inhibited the ERK/STAT3 pathway and activated the E-cadherin pathway in human and murine cell lines (28). ...
Article
Full-text available
This review presents an in-depth analysis of the immense potential of CRISPR-Cas9 technology in revolutionizing oral cancer research. It underscores the inherent limitations of conventional treatments while emphasizing the pressing need for groundbreaking approaches. The unparalleled capability of CRISPR-Cas9 to precisely target and modify specific genes involved in cancer progression heralds a new era in therapeutic intervention. Employing genome-wide CRISPR screens, vulnerabilities in oral cancer cells can be identified, thereby unravelling promising targets for therapeutic interventions. In the realm of oral cancer, the disruptive power of CRISPR-Cas9 manifests through its capacity to perturb genes that are intricately associated with drug resistance, consequently augmenting the efficacy of chemotherapy. To address the challenges that arise, this review diligently examines pertinent issues such as off-target effects, efficient delivery mechanisms, and the ethical considerations surrounding germline editing. Through precise gene editing, facilitated by CRISPR/Cas9, it becomes possible to overcome drug resistance by rectifying mutations, thereby enhancing the efficacy of personalized treatment strategies. This review delves into the prospects of CRISPR-Cas9, illuminating its potential applications in the domains of medicine, agriculture, and biotechnology. It is paramount to emphasize the necessity of ongoing research endeavors and the imperative to develop targeted therapies tailored specifically for oral cancer. By embracing this comprehensive overview, we can pave the way for ground-breaking treatments that instill renewed hope for enhanced outcomes in individuals afflicted by oral cancer.
Article
Full-text available
With the lack of specific signs and symptoms, pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at late metastatic stages, resulting in poor survival outcomes. Among various biomarkers, microRNA-21 (miR-21), a small non-coding RNA, is highly expressed in PDAC. By inhibiting regulatory proteins at the 3′ untranslated regions (UTR), miR-21 holds significant roles in PDAC cell proliferation, epithelial–mesenchymal transition, angiogenesis, as well as cancer invasion, metastasis, and resistance therapy. We conducted a systematic search across major databases for articles on miR-21 and pancreatic cancer mainly published within the last decade, focusing on their diagnostic, prognostic, therapeutic, and biological roles. This rigorous approach ensured a comprehensive review of miR-21’s multifaceted role in pancreatic cancers. In this review, we explore the current understandings and future directions regarding the regulation, diagnostic, prognostic, and therapeutic potential of targeting miR-21 in PDAC. This exhaustive review discusses the involvement of miR-21 in proliferation, epithelial–mesenchymal transition (EMT), apoptosis modulation, angiogenesis, and its role in therapy resistance. Also discussed in the review is the interplay between various molecular pathways that contribute to tumor progression, with specific reference to pancreatic ductal adenocarcinoma.
Conference Paper
Full-text available
https://www.hilarispublisher.com/proceedings/the-wonders-of-crispr-to-cancer-therapy-36856.html
Article
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer that is strongly associated with poor prognosis and short median survival times. In stark contrast to the progress seen in other cancer types in recent decades, discoveries of new treatments in PDAC have been few and far between and there has been little improvement in overall survival (OS). The difficulty in treating this disease is multifactorial, contributed to by late presentation, difficult access to primary tumour sites, an ‘immunologically cold’ phenotype, and a strong tendency of recurrence likely driven by cancer stem cell (CSC) populations. Furthermore, apparently contrasting roles of tumour components (such as fibrotic stroma) and intracellular pathways (such as autophagy and TGFβ) have made it difficult to distinguish beneficial from detrimental drug targets. Despite this, progress has been made in the field, including the determination of mFOLFIRINOX as the standard-of-care adjuvant therapy and the discovery of KRASG12C mutant inhibitors. Moreover, new research, as outlined in this review, has highlighted promising new approaches including the targeting of the tumour microenvironment, enhancement of immunotherapies, epigenetic modulation, and destruction of CSCs.
Article
Full-text available
CRISPR/Cas9 is a promising technology for gene editing. To date, intracellular delivery vehicles for CRISPR/Cas9 are limited by issues of immunogenicity, restricted packaging capacity, and low tolerance. Here, we report an alternative, nonviral delivery system for CRISPR/Cas9 based on engineered exosomes. We show that non-autologous exosomes can encapsulate CRISPR/Cas9 plasmid DNA via commonly available transfection reagents and can be delivered to recipient cancer cells to induce targeted gene deletion. As a proof-of-principle, we demonstrate that exosomes loaded with CRISPR/Cas9 can target the mutant Kras G12D oncogenic allele in pancreatic cancer cells to suppress proliferation and inhibit tumor growth in syngeneic subcutaneous and orthotopic models of pancreatic cancer. Exosomes may thus be a promising delivery platform for CRISPR/Cas9 gene editing for targeted therapies.
Article
Full-text available
Cancer is associated with single or multiple gene defects. Recently, much research has focused on incorporating genetic materials as one of the means to treat various types of carcinomas. RNA interference (RNAi) conveys an alternative genetic approach for cancer patients, especially when conventional medications fail. RNAi involves the inhibition of expression of specific messenger RNA that signals for uncontrollable cell growth and proliferation, most notably with carcinoma cells. This molecular technology is promising as genetic materials allow us to overcome issues associated with chemotherapeutic agents including organ damage associated with severe drug toxicities. Nonetheless, vast challenges impede successful gene therapy application, including low tumor localization, low stability and rapid clearance from the blood circulation. Owing to the limited treatment opportunities for the management of cancer, the development of effective siRNA carrier systems involving nanotherapeutics has been extensively explored. Over the past years, several siRNA nanotherapeutics have undergone a period of clinical investigation, with some demonstrating promising antitumor activities and safety profiles. Extensive observation of siRNA-nanoparticles is necessary to ensure commercial success. Therefore, this review mainly focuses on the progress of siRNAs-loaded nanoparticles that have undergone clinical trials for cancer treatment. The status of the siRNA nanotherapeutics is discussed, allowing comprehensive understanding of their gene-mediated therapeutics.
Article
Full-text available
The serious drawbacks of the conventional treatment of pancreatic ductal adenocarcinoma (PDAC) such as nonspecific toxicity and high resistance to chemo and radiation therapy, have prompted the development and application of countless small interfering RNA (siRNA)‐based therapeutics. Recent advances in drug delivery systems hold great promise for improving siRNA‐based therapeutics and developing a new class of drugs, known as nano‐siRNA drugs. However, many fundamental questions, regarding toxicity, immunostimulation, and poor knowledge of nano‐bio interactions, need to be addressed before clinical translation. In this review, we provide recent achievements in the design and development of various nonviral delivery vehicles for pancreatic cancer therapy. More importantly, codelivery of conventional anticancer drugs with siRNA as a new revolutionary pancreatic cancer combinational therapy is completely discussed.
Article
Full-text available
Song Zhang, 1, 2 Zhujun Cheng, 3 Yanan Wang, 1 Tianyu Han 1 1Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China; 2College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, People’s Republic of China; 3Department of Burn, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of ChinaCorrespondence: Tianyu Han Email hantianyu87@163.comAbstract: RNAi therapeutics have been growing. Patisiran and givosiran, two siRNA-based drugs, were approved by the Food and Drug Administration in 2018 and 2019, respectively. However, there is rare news on the advance of miRNA drugs (another therapeutic similar to siRNA drug). Here we report the existing obstacles of miRNA therapeutics by analyses for resources available in a drug target perspective, despite being appreciated when it began. Only 10 obtainable miRNA drugs have been in clinical trials with none undergoing phase III, while over 60 siRNA drugs are in complete clinical trial progression including two approvals. We mechanically compared the two types of drug and found that their major distinction lay in the huge discrepancy of the target number of two RNA molecules, which was caused by different complementary ratios. One miRNA generally targets tens and even hundreds of genes. We named it “too many targets for miRNA effect” (TMTME). Further, two adverse events from the discontinuation of two miRNA therapeutics were exactly answered by TMTME. In summary, TMTME is inevitable because of the special complementary approach between miRNA and its target. It means that miRNA therapeutics would trigger a series of unknown and unpreventable consequences, which makes it a considerable alternative for application.Keywords: RNAi, miRNA, siRNA, therapeutics
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers worldwide due to its aggressiveness and the challenge to early diagnosis and treatment. In recent decades, nanomaterials have received increasing attention for diagnosis and therapy of PDAC. However, these designs are mainly focused on the macroscopic tumor therapeutic effect, while the crucial nano–bio interactions in the heterogeneous microenvironment of PDAC remain poorly understood. As a result, the majority of potent nanomedicines show limited performance in ameliorating PDAC in clinical translation. Therefore, exploiting the unique nature of the PDAC by detecting potential biomarkers together with a deep understanding of nano–bio interactions that occur in the tumor microenvironment is pivotal to the design of PDAC‐tailored effective nanomedicine. This review will introduce tailor‐made nanomaterials‐enabled laboratory tests and advanced noninvasive imaging technologies for early and accurate diagnosis of PDAC. Moreover, the fabrication of a myriad of tailor‐made nanomaterials for various PDAC therapeutic modalities will be reviewed. Furthermore, much preferred theranostic multifunctional nanomaterials for imaging‐guided therapies of PDAC will be elaborated. Lastly, the prospects of these nanomaterials in terms of clinical translation and potential breakthroughs will be briefly discussed.
Article
Full-text available
CRISPR gene editing technology belongs to the third generation of gene editing technology. Since its discovery, it has attracted the attention of a large number of researchers. Investigators have published a series of academic articles and obtained breakthrough research results through in-depth research. In recent years, this technology has developed rapidly and been widely applied in many fields, especially in medicine. This review focuses on concepts of CRISPR gene editing technology, its application in cancer treatments, its existing limitations, and the new progress in recent years for detailed analysis and sharing.
Article
It is challenging to diagnose patients with pancreatic ductal adenocarcinoma (PDAC) early on, and their treatment is often complex. Gemcitabine (GEM) is the first-line treatment for PDAC, but its efficacy is limited in most patients due to the GEM resistance from KRAS and P53 gene mutations. We describe the correction of a double gene mutation and therapeutic effect for the GEM resistant PDAC. Bio-available nanoliposomes (NL) possessing Cas9-ribonucleoproteins and adenine-base editors were developed to conduct KRAS and P53 mutation gene editing directly. NLs were conjugated with EGFR antibodies to tumor-specific delivery, and the anti-cancer effect was verified in vitro and in vivo Model. Our GEM-combinatorial therapeutic strategies using double gene editing systems with one-shot may be a potent therapy for PDAC, overcoming chemoresistance.
Article
Importance Pancreatic ductal adenocarcinoma (PDAC) is a relatively uncommon cancer, with approximately 60 430 new diagnoses expected in 2021 in the US. The incidence of PDAC is increasing by 0.5% to 1.0% per year, and it is projected to become the second-leading cause of cancer-related mortality by 2030. Observations Effective screening is not available for PDAC, and most patients present with locally advanced (30%-35%) or metastatic (50%-55%) disease at diagnosis. A multidisciplinary management approach is recommended. Localized pancreas cancer includes resectable, borderline resectable (localized and involving major vascular structures), and locally advanced (unresectable) disease based on the degree of arterial and venous involvement by tumor, typically of the superior mesenteric vessels. For patients with resectable disease at presentation (10%-15%), surgery followed by adjuvant chemotherapy with FOLFIRINOX (fluorouracil, irinotecan, leucovorin, oxaliplatin) represents a standard therapeutic approach with an anticipated median overall survival of 54.4 months, compared with 35 months for single-agent gemcitabine (stratified hazard ratio for death, 0.64 [95% CI, 0.48-0.86]; P = .003). Neoadjuvant systemic therapy with or without radiation followed by evaluation for surgery is an accepted treatment approach for resectable and borderline resectable disease. For patients with locally advanced and unresectable disease due to extensive vascular involvement, systemic therapy followed by radiation is an option for definitive locoregional disease control. For patients with advanced (locally advanced and metastatic) PDAC, multiagent chemotherapy regimens, including FOLFIRINOX, gemcitabine/nab-paclitaxel, and nanoliposomal irinotecan/fluorouracil, all have a survival benefit of 2 to 6 months compared with a single-agent gemcitabine. For the 5% to 7% of patients with a BRCA pathogenic germline variant and metastatic PDAC, olaparib, a poly (adenosine diphosphate [ADB]-ribose) polymerase inhibitor, is a maintenance option that improves progression-free survival following initial platinum-based therapy. Conclusions and Relevance Approximately 60 000 new cases of PDAC are diagnosed per year, and approximately 50% of patients have advanced disease at diagnosis. The incidence of PDAC is increasing. Currently available cytotoxic therapies for advanced disease are modestly effective. For all patients, multidisciplinary management, comprehensive germline testing, and integrated supportive care are recommended.