You are on page 1of 26

Pharm Res

DOI 10.1007/s11095-017-2134-2

EXPERT REVIEW

The Race of 10 Synthetic RNAi-Based Drugs


to the Pharmaceutical Market
Ricardo Titze-de-Almeida 1 & Catherine David 2 & Simoneide Souza Titze-de-Almeida 1

Received: 22 November 2016 / Accepted: 27 February 2017


# Springer Science+Business Media New York 2017

ABSTRACT Ten years after Fire and Melo’s Nobel Prize for biological barriers), RNAi definitively opens a wide avenue for
discovery of gene silencing by double-stranded RNA, a re- drug development.
markable progress was achieved in RNA interference
(RNAi). Changes in the chemical structure of synthetic oligo- KEY WORDS miRNA . pharmaceutical market . RNA
nucleotides make them more stable and specific, and new interference . RNAi . siRNA
delivery strategies became progressively available. The atten-
tion of pharmaceutical industry rapidly turned to RNAi, as an
opportunity to explore new drug targets. This review ad- ABBREVIATIONS
dresses nine small-interfering RNAs (siRNAs) and one unique LNA Locked nucleic acid
microRNA (miRNA) inhibitor, which entered the phase 2–3 LNP Lipid nanoparticle
clinical trials. The siRNAs in focus are PF-04523655, TKM- LODER LOcal Drug EluteR
080301, Atu027, SYL040012, SYL1001, siG12D-LODER PEG Polyethylene glycol
(phase 2), QPI-1002, QPI-1007, and patisiran (phase 3). PLGA poly(lactic-co-glycolic) acid
Regarding miRNAs, their content can be down- or up-regu- RISC RNA-induced silencing complex
lated, by using miRNA inhibitors (AntimiRs) or miRNA RNAi RNA interference
mimics. Miravirsen is an AntimiR-122 for hepatitis C virus SNALPs Stable nucleic acid lipid particles
infection. The flexibility of RNAi technology is easily under-
stood taking into account: (i) the different drug targets (i.e.
p53, caspase 2, PKN3, β2-adrenergic receptor, mutated INTRODUCTION
KRAS, microRNAs); (ii) therapeutic conditions, including
ophthalmic diseases, kidney injury, amyloidosis, pancreatic RNA interference (RNAi) therapeutics comprise a relatively
cancer, viral hepatitis; and (iii) routes of administration (ocu- new class of drugs that are about to reach the pharmaceutical
lar, intravenous, subcutaneous, intratumoral). Although some market. All these drugs present a chemical structure of short
issues are still matters of concern (delivery, toxicity, cost, and chains of nucleotides that essentially differ in their sequence of
puric and pyrimidic bases (1).
While proteins are the most common targets for traditional
medicines like β-adrenergic antagonists for glaucoma treat-
* Ricardo Titze-de-Almeida ment, the messenger RNA (mRNA) is the target of RNAi-
ricardotitze.unb@gmail.com based drugs. In this sense, siRNA SYL040012 will counteract
β2-adrenergic activity by cleaving its mRNA, thus reducing
Catherine David
http://biotika.com.br
receptors that stimulate aqueous humor production in the
eye (2). RNAi, post-transcriptional gene silencing or knock
1
Technology for Gene Therapy Laboratory, ASS 128, ICC Sul, University
down are synonymous for a process of mRNA downregula-
of Brasília–UnB, Campus Darcy Ribeiro, FAV., Brasília, DF 70910-970, tion that directly affects the respective protein level (Box I) (3,
Brazil 4). Proteins comprise the vast majority of pharmacological
2
Av. Pedro Severino Junior, 366, sala 45 e 46 – Vila Guarani, São drug targets, meaning that RNAi holds an inherent broad
Paulo, SP 04310-060, Brazil therapeutic potential. Finding the correct target for knock
Titze-de-Almeida, David and Titze-de-Almeida

down is the first technological challenge; delivery is the sec- PF-04523655 injected by IVT route shows plasmatic levels
ond, not the less important. within a few hours (18).
Many aspects of RNAi, including origins, cell pathways, PF-04523655 was well-tolerated in preclinical tests with
and biological roles are issues revised elsewhere (5–8). This rats and cynomolgus monkeys (18). Local adverse effects were
review focuses on ten synthetic molecules that take advantage only detected at the highest IVT injection of 3 mg/eye in
of RNAi pathways, which are shortly described in Fig. 1. Nine monkeys. The first phase 1 human clinical trial
of them are small-interfering RNAs (siRNAs) that are du- (NTC00725686) included subjects that received increasing
plexes of 19–23 ribonucleotides with sense (passenger) and doses in single IVT injection of PF 04523655 and were follow-
antisense (guide) strands (Fig. 2a). The duplex is incorporated ed up for 24 months. This dose-escalation study evaluated the
into a complex of proteins named RNA-induced silencing safety, tolerability, pharmacokinetics, and dose-limiting toxic-
complex (RISC) that will first remove the passenger strand. ities. For safety reasons, the study was organized in two strata
A seed region of the remaining guide strand will look for with specific end points. In the first one, the drug was admin-
complementary nucleotides in mRNA sequence and bind to istered to individuals legally blind (21) at doses of 0.05–3 mg
them by hydrogen bonds (9, 10). Then RISC complex will (n = 3 individuals per dose). They presented neovascular
execute cleavage of the mRNA found by the guide strand AMD unresponsive to prior treatment and a best corrected
(Fig. 1a) (11, 12). Finally, a single RNAi drug is a microRNA visual acuity (BCVA) ≤ 20/200. After the first safety results of
inhibitor to miR-122 (AntimiR-122). AntimiRs are antisense PF-04523655 in phase 1, regulatory authorities approved fur-
oligodeoxynucleotides targeted to specific miRNAs, thus ther trials and the sponsor tested efficacy in phase 2. It includ-
abolishing its role in disease pathogenesis (see Fig. 1b). ed 27 patients with potential to benefit from therapy using
The present work reviews preclinical and clinical studies on doses ≥1 mg from end point 1; BCVA between 20/200 and
ten synthetic RNAi-based drugs that entered phase 2–3 of 20/800. For dose-escalation, individuals received the next
human clinical trials registered in U.S. National Institutes of higher dose of PF-04523655 provided no dose-limiting toxic-
Health - NIH (Table I). The small-interfering RNAs (siRNAs) ity had been detected. The study found at least one adverse
are PF-04523655, QPI-1002, QPI-1007, TKM-080301, event in ≈ 70% of individuals, but no dose-limiting toxicity.
Atu027, SYL040012, SYL1001, patisiran, and siG12D- Undesired reactions were mild to moderate and not related to
LODER. A microRNA inhibitor for hepatitis C virus infec- PF-04523655. Only one report of retinal pigment epithelial
tion named miravirsen is also part of this review. These oligo- detachment could possibly be related to PF-04523655
nucleotides are candidates to inaugurate a new class of thera- (1.5 mg), in a patient that started receiving ranibizumab in
peutic agents in the pharmaceutical market. the study eye. Part of the drug injected into the eye was
absorbed into systemic circulation. IVT injection of PF-
04523655 in doses ≥0.4 mg produced detected plasma levels
at 1, 4, and 24 h post injection. However, all tested doses were
SIRNAS IN CLINICAL STUDIES REGISTERED IN below the lowest limit of quantification by day 14. The max-
NIH imal concentration in plasma was between 1–4 h after injec-
tion. The highest median plasma concentration was 2.86 ng/
Pf-04523655 (Pf-655) mL at 4 h after IVT injection of 2.25 mg of PF-04523655. In
summary, this study shows that a single IVT injection of PF-
PF-04523655 is a siRNA designed to silence RTP801 in indi- 04523655 ≤ 3 mg was safe and well-tolerated in patients
viduals with neovascular age-related macular degeneration with AMD. Although the main focus of this phase 1
(AMD). Patients with choroidal neovascularization secondary study was toxicity, it also evaluated a possible efficacy
to AMD and diabetic retinopathy overexpress RTP801 as an of PF-04523655 in the end point 2. Unfortunately, a
hypoxia-inducible gene, which plays a role in disease patho- single IVT injection of PF-04523655 had no benefit in
genesis (13–15). Regarding siRNA structure, PF-04523655 is visual acuity or retinal thickness (18).
a duplex of 19 ribonucleotides, stabilized by 2′-O-methylation The first phase 2 clinical trial on PF-04523655, the Monet
that introduces a 2′-methoxy group in ribose (2′-O-Me), as study (NCT00713518), evaluated the efficacy of PF-
shown in Fig. 2b (16, 17). Studies of RNA silencing were 04523655 in monotherapy and in association with
performed by intravitreal (IVT) injection (18). It is the same ranibizumab for AMD treatment (16). All individuals first re-
procedure as for ranibizumab (Lucentis®), a standard therapy ceived ranibizumab 0.5 mg at a baseline. The study tested the
for AMD based on the inhibition of vascular endothelial following conditions: 1. Two doses of PF-04523655 in mono-
growth factor (VEGF) (19). Even being a naked siRNA, PF- therapy, every 4 weeks for 8 weeks (1 mg or 3 mg); 2. PF-
04523655 showed the ability to penetrate retinal cells without 04523655 in monotherapy, every 2 weeks for 8 weeks
help of any transfection reagent and to silence RTP801 in (3 mg); 3. An association of PF-04523655 + ranibizumab
assays in vitro and in vivo (20). Also in a pharmacokinetic study, (0.5 mg), every 4 weeks for 8 weeks (1 mg or 3 mg). The
RNAi-based drugs in phase II – III clinical trials

Fig. 1 Synthetic RNAi-based drugs modulate RNAi pathways. (a) siRNA pathway. First, double-stranded RNA (dsRNA) molecules from infecting pathogens or
cellular genes are processed by a ribonuclease enzyme Dicer, which interacts with TRBP and PACT, forming short dsRNA with ≈ 22 nucleotides and two
nucleotide overhangs at 3' end – siRNAs. Synthetic siRNAs can also be introduced into cells (red box). Regardless their origin, siRNAs will be incorporated into a
complex of proteins: RNA-induced silencing complex (RISC). The endonuclease Argonaute 2 (AGO) removes passenger (sense) strand of the siRNA, allowing
antisense or guide strand to bind to fully complementary sequences in the mRNA coding region (CDS). In summary, siRNAs conduct the active RISC to specific
mRNA for cleavage by AGO enzyme. (b) miRNA pathway. MicroRNAs (miRNA) are coded by genes present in mammalian DNA. At the top, RNA polymerase
II (Pol II, in brown) will transcribe miRNA genes in long RNA sequences of hundreds of nucleotides. Transcripts will form a peculiar shape of a hairpin. Each hairpin
is a duplex of ≈ 70 partially complementary ribonucleotides that ends with a short non-complementary sequence (loop). Except for the hairpin, miRNA has an
mRNA shape. miRNA is processed in cell nucleus where it is 7-methyl-guanosine capped (m7G, filled red circle) in 5' end and polyadenylated at 3' end, forming
the primary miRNA (Pri-miRNA). Drosha and DGCR8 enzymes will cleave RNA sequences outside the hairpin to form miRNA precursor (Pre-miRNA), further
exported to cytoplasm by Exportin 5. As for siRNA pathway above described, Dicer enzyme will process the Pre-miRNA into a miRNA with 18 – 25 nucleotides.
The miRNA duplex associates with the RISC (miRISC), sense strand is removed, and the guide strand binds complementary sequences in target mRNAs. The
difference with siRNA is that miRNAs binds to several mRNAs with partially complementary pairing, and target sequences are placed in 3'-UTR region. The
absence of a full complementarity between miRNA and mRNA sequences compromises AGO endonuclease activity. In consequence, effect is a translational
repression or mRNA degradation instead of cleavage of mRNA by AGO. Synthetic miRNAs (miRNA mimic) or antisense sequences that inhibit specific miRNAs
(AntimiR) will artificially modulate the miRNA content (orange boxes). AGO Argonaute 2, CDS Coding sequence region of mRNA, DGCR8 DiGeorge syndrome
critical region gene 8, Dicer a ribonuclease enzyme, Drosha a ribonuclease enzyme, miRISC miRNA duplex associated with the RISC complex, PACT Protein
activator of the interferon-induced protein kinase (PKR), Pol II RNA polymerase II, RISC RNA-induced silencing complex, TRBP HIV-1 Trans-activation response
(TAR) RNA-binding protein.

control group was treated with only ranibizumab (0.5 mg). All in comparison with ranibizumab alone (6.8 letters). This asso-
monotherapy protocols with PF-04523655 were less effective ciation, however, was not superior to ranibizumab in reducing
than ranibizumab. Although not statistically significant the central subfield retinal thickness and total subfoveal cho-
(P = 0.33), treatment with PF-04523655 + ranibizumab roidal neovascularization (CNV) area. The same occurred for
showed a higher improvement of BCVA values (9.5 letters) decreases in lesion thickness. Finally, some adverse reactions
Titze-de-Almeida, David and Titze-de-Almeida

Fig. 2 Schematic structure of a siRNA duplex and chemical modifications in oligonucleotides. (a) A typical 21-mer siRNA with its annealed sense and antisense
strands, and two overhangs nucleotides. RNA duplex is dismounted in cell cytoplasm by RISC complex, and the sense strand is eliminated. The antisense strand
will guide the RISC complex to the target mRNA for cleavage. (b) Schematic changes in RNA-backbone. At the center, an unmodified ribose for comparisons.
Letter B represents a nucleotide base. Ribose modifications ring include fluorine (2'-F), methoxy (2'-OMe), locked nucleic acids (LNA) and unlocked nucleic acids
(UNA). Phosphodiester bond can be substituted by phosphorothioate. Chemical modifications of siRNA nucleotides improves nucleases resistance and reduces
immunogenicity and off-target silencing, not affecting efficacy.

not related to PF-04523655, mild to moderate in severity, also reduces costs. A significant limitation of PF-04523655 is
occurred in the study eye (4). efficacy as the siRNA was not superior to the reference drug
Matisse study was the third phase 2 clinical trial and fo- ranibizumab (anti-VEGF) in patients with AMD. PF-
cused on Diabetic Macular Edema - DME (NCT01445899). 04523655 has only shown a tendency to increase the visual
This study evaluated the efficacy and safety of PF-04523655, acuity when administered in combination with ranibizumab,
in monotherapy or in association with ranibizumab. It was without improving eye disease histopathology. Despite the
organized in two strata. The first analyzed safety, dose- interest of Pfizer that get a license for the technology, it may
limiting toxicity, and pharmacokinetics of a single IVT injec- be difficult to convince reimbursement organizations to pay
tion of PF-04523655 in subjects with low vision. The second for, as a current standard treatment for AMD gives reasonably
evaluated the safety, tolerability, and ability to improve visual good results. However, the use of PF-04523655 can be fa-
acuity. For the latter, patients with DME received 6 monthly vored whether it is confirmed that anti-VEGF agents present
IVT injections of PF-04523655 alone or associated with a potential risk for geographic atrophy progression in patients
ranibizumab. Anatomical changes in the retina and retinal with AMD (22–24). Finally, there is an expectation about a
nerve fiber layer morphology were also examined. This study phase 2 study on PF-04523655 for patients with another eye
is completed, but no results have been published yet. disorder, DME.

Critical Analysis QPI-1002 (I5NP)

PF-04523655 uses the intravitreal route of administration, a QPI-1002 was designed to protect proximal tubule cells in
standard procedure as for ranibizumab. This route will re- kidney injury associated with ischemia or nephrotoxicity.
quire an ophthalmologist assistance for the dosing regimen, Acute kidney injury (AKI) is a new term for acute renal failure,
every 2–4 weeks for eight weeks, or 6 monthly injections. a clinical syndrome characterized by a rapid decrease in renal
Regional administration typically brings a significant advan- excretory function (25). AKI is an important health problem
tage: the dose is reduced making the treatment cheaper. PF- due the incidence, mortality rate, and risk for patients to de-
04523655 in fact required 1 or 3 mg per administration, an velop a chronic and dialysis dependent disease (26, 27). The
amount relatively small compared to other drugs of this re- death of renal cells by apoptosis plays an important role in
view. Indeed, it was not formulated in nanoparticles which AKI pathogenesis, regardless its etiology was ischemia, sepsis,
RNAi-based drugs in phase II – III clinical trials

or a nephrotoxic injury (28). Thus, the inhibition of apoptotic via tail vein. Sub-acute toxicity of QPI-1002 (10 mg/Kg) was
pathways in injured renal cells would be of value in AKI. The also tested in nephrectomized rats, as a model of renal impair-
siRNA in focus, QPI-1002, was designed to silence the pro- ment. Acute toxicity test in rats included single I.V. injections
apoptotic p53 protein in proximal tubule cells (PTC), thus of QPI-1002 (200, 800, 1200 or 2000 mg/Kg) or QM5
reducing the ischemia-reperfusion injury. It is a naked, (1200 mg/Kg). Finally, the study evaluated QPI-1002 in ge-
blunt-ended 19-base-pair duplex, and 2′-O methylated (29). netic toxicity assays. Plasma concentration of QPI-1002 after
QPI-1002 was the first siRNA systemically administered to I.V. injection decreased rapidly, >90% in 30 min post-dosing
humans, given by the intravenous route (I.V.). Intravenously (200 mg/Kg). The drug was predominantly distributed to the
injected siRNAs have a peculiar pharmacokinetics: they are kidney, as expected. This finding agrees with in vivo imaging by
internalized mainly by kidney and liver cells (30). This pattern two-photon microscopy that showed fluorescent labeled
of distribution was confirmed in various studies with RNA siRNAs in kidney tissue. About renal cells, QPI-1002 accumu-
duplexes and, in addition, with single-stranded antisense lates mainly in PTC, with a peak at 5–30 min post-injection.
DNA oligonucleotides (31–35). As the kidney is a natural tar- The levels decreased in 2 h, and became undetectable after
get for I.V. injected oligonucleotides, kidney diseases caught 24 h. This data agreed with a previous work that found a
the attention for siRNA-based therapeutics. decrease of p53 mRNA content in PTC for 24–48 h post-
Molitoris et al. (2009) executed the first preclinical study on injection of QPI-1002 (29). The temporary and site-specific
QPI-1002, testing its efficacy in ischemic and chemotoxic effect of this siRNA is a valuable strategy to control the apo-
models of AKI. First, the potential reduction of ischemic inju- ptosis of tubular cells soon after an acute injury. Afterwards,
ry of kidneys was evaluated in rats submitted to bilateral renal- p53 expression will return to the basal level and cells with
clamps. Animals received I.V. injections of 1 mg/Kg each of irreversible damage could be removed. QPI-1002 caused no
QPI-1002, before (−2 h, −30 min) and after (+4 h, + 8 h) the adverse effects on the cardiovascular, respiratory and central
injury (29). Following the expected pattern of distribution, nervous systems. Toxicity was found only for doses ≥800 mg/
QPI-1002 concentrated in PTC cells. Serum creatinine levels Kg in rats and ≥1000 mg/Kg in monkeys. QPI-1002 was not
of ischemic rats decreased from 3.7 mg/dL to 1.9 mg/dL in genotoxic.
the siRNA-treated group, showing the improvement of renal The first human phase 1 clinical trial on QPI-1002
function (p < 0.01). Apoptosis of PTC cells caused by ischemia (NCT00554359) was carried on patients undergoing cardio-
was reduced. In addition, this p53-targeted siRNA attenuated vascular surgery. Those individuals received a single I.V. in-
kidney injury in the model of aortic clamp. Rats have received jection of QPI-1002 at 4 h after removal of the cardiopulmo-
QPI-1002 via I.V. 4 h after a 60 min of 90% suprarenal aortic nary bypass machine. No results are available but the drug
clamp. QPI-1002 also protected renal function from the inju- had to be well-tolerated, as the study advanced to a phase 2
ry caused by the chemotoxic agent cisplatin (7.5 mg/Kg, in- clinical trial (NCT02610283). The study will evaluate the ef-
traperitoneal route). For that, the animals received three ficacy and safety of QPI-1002 in patients (n = 340) who are at
siRNA injections (12 mg/ Kg), 30 min before and on high risk for AKI following cardiac surgery. This clinical trial
days two and three after cisplatin administration (29). is now recruiting participants.
Indeed, QPI-1002 showed efficacy in rats subjected to QPI-1002 could also protect renal cells in patients that
renal transplantation (36). receive kidney transplants. Those patients can develop a form
An extensive pharmacokinetic and toxicological testing on of AKI related to the transplant and named delayed graft
QPI-1002 was executed in rat and nonhuman primates to function (DGF) (38). It occurs due the ischemia-reperfusion
support the first-in-man clinical trial (37). The study first used injury, and may result in renal damage. Patients will show
naive male cynomolgus monkeys. They received 4 separate post-transplantation oliguria, with an increased risk for allo-
I.V. doses of QPI-1002, at Day 4 (5 mg/Kg/dose) and Day 11 graft rejection and lower long-term survival (39). Thus, Quark
(25 mg/Kg/dose). The authors opted for 4 doses to provide performed a clinical trial phase 1/2 with patients undergoing
flexibility for further tests in human clinical trials. Animals deceased donor kidney transplantation (NCT00802347).
were examined for general health, food intake, cardiovascu- Besides drug safety evaluation in phase 1, the study also ex-
lar-, respiratory-, and nervous system activity, as well as serum amined whether QPI-1002 would protect transplanted pa-
chemistry and hematology. For sub-acute toxicity, monkeys tients from DGF. The clinical trial had two main arms. Part
received 3, 12, or 48 mg/Kg/dose by I.V. route (brachial A was a dose-escalation study to find out the maximum toler-
veins). For acute toxicity, animals received a single injection ated dose of QPI-1002 (0.5 mg/Kg, 1.5 mg/Kg, 5 mg/Kg, or
of QPI-1002 (200, 500, or 1000 mg/Kg). This study also 10 mg/Kg). Part B evaluated safety and efficacy of QPI-1002,
tested QPI-1002 in rats, as a second species, for sub-acute at the dose 10 mg/Kg identified in part A. In this phase 2
and acute toxicity and pharmacokinetics. In sub-acute toxicity study (NCT00802347, QRK.006B), the drug was adminis-
tests, the animals received QPI-1002 (3, 15, or 60 mg/Kg/ tered in a single I.V. dose, at 30 min after reperfusion.
dose) or a rat analogous named QM5 (15 or 60 mg/Kg/dose) Individuals were organized in groups, according to the donor
Titze-de-Almeida, David and Titze-de-Almeida

and method for tissue preservation. Patients (n = 331) were Novartis has invested 10 million USD for development and
from 52 transplant centers in North America and Europe (40). commercialization of QPI-1002 (41).
The relative risk of DGF was defined as the need for dialysis
within the first 7 days post-transplant, excluding dialysis in the QPI-1007
first 24 h because of hyperkalemia/hypervolemia. As cited
above, patients received a single I.V. injection of QPI 1002 The loss of retinal ganglion cells (RGC) is a hallmark of
(10 mg/Kg) at 30 min after reperfusion. The intended end ophthalmic diseases that result in visual impairment and
point of a 30% decrease in relative risk of DGF in QPI-1002- blindness like nonarteritic anterior ischemic optic neu-
treated individuals was not achieved for the total population, ropathy - NAION and glaucoma (42–44). As apoptosis
as it reached 15%. However, QPI-1002 could reduce by is a key event in RGC loss, proteins involved in apo-
30.5% this relative risk of DGF in patients of the group named ptotic pathways are attractive targets for therapeutics.
ECD/CS (expanded criteria donor/cold-stored) (n = 177). A Caspase-2 is a protein expressed by RGC during injury,
similar effect occurred in patients that received kidney grafts and treatments which inhibit this pro-apoptotic protein
from older donors (˃ 35 or 45 years old). Positive changes in have provided neuroprotective effect (45–47). QPI-1007
dialysis after transplant was another benefit of QPI-1002, is a caspase-2-targeted siRNA developed to protect
found in ECD/CS group. It extended the time for the first RGC in optic neuropathies, by IVT injection (48). It
dialysis in the first post-transplant month. QPI-1002-treated is a naked RNA duplex of 19 nucleotides, with 2′-O-
patients also showed a lower number of dialysis sessions (6.0 vs. methylated sugar nucleosides in the guide strand.
11.2) and a shorter first course of dialysis (13.4 vs. 25.3 days). Passenger strand contains chemical changes at both
Finally, patients treated with QPI-1002 showed a tendency to ends, as shown in Fig. 3a. One L-DNA cytidine nucle-
need 1.5 fold less dialysis sections in the 6-month study period otide placed before the last nucleotide at 3´ end, and an
of follow up (375 vs. 561, p = 0.059). Regarding safety, Quark inverted deoxyabasic moiety at 5′ end, represented by
reported a similar safety profile between QPI-1002-treated the letters BY^ and Bia^, respectively (49). QPI-1007
and placebo groups (40). In 2010, the company established guide strand will form Watson & Crick’s base pairs with
an agreement with Novartis for the development and com- nucleotides 240–258 of human caspase-2 mRNA se-
mercialization of QPI-1002 (41). quence (transcript variant 2, NM_001224.4; Fig. 3b), a
Quark is recruiting patients for a phase III clinical trial on key step in the siRNA pathway explained in Fig. 1. The
QPI-1002 (NCT02610296, QRK306), with an estimated en- rat orthologous mRNA has an identical 5′-
rollment of 634 individuals. The ReGIFT study will evaluate gccagaatgtggaactcct −3′ target sequence in positions
whether this drug reduces DGF in patients that receive kidney 251–269 (NM_022522.2; Fig. 3b). It allowed the use
allografts from donors ˃ 45 years after brain death. of identical siRNAs in both species.
QPI-1007, then named siCASP2, has shown ability to si-
lence caspase-2 expression in cells in culture (48). It caused
Critical Analysis 80% and 65% knock-down of caspase-2 mRNA in human
Hela cells and rat PC12 cells, respectively. siCASP2 resisted
QPI-1002 addresses AKI, a severe renal disease that may to nuclease attack in serum and vitreous humor during 24 h at
require dialysis - a significant constraint and expensive pro- 37°C, without causing immune or off target effects. Most
cess. The therapeutic rationale is to suppress the pro-apoptotic RGC had internalized siCASP2 in few hours, as revealed by
p53 protein to reduce the death of renal cells, thus preserving Cy3-labelled siRNAs. Finally, the most important result –
the kidneys physiology. The intended endpoint of 30% reduc- siCASP2 reduced RGC apoptosis in two rat models of acute
tion in DGF - a form of AKI related to transplants, was met optic nerve (ON) injury. In the model named ONC (optic
for a subgroup of individuals and not the entire population. nerve crushed), the ON of both eyes are crushed infraorbital
Indeed, QPI-1002 showed a tendency to reduce the need for to transect all RGC axons, causing a high cell loss at 7 days.
dialysis in patients with kidney transplants. Thus efficacy is a siCASP2 caused a significant dose-dependent protection of
critical point for QPI-1002. In contrast, this siRNA holds RGC at doses of 5, 10 and 20 μg per eye. Also, doses of 20
many positive properties: (i) as a naked siRNA, it can be a and 35 μg keep RGC density in injured eyes to values close to
relatively cheaper drug; (ii) treatment is via I.V. route, com- intact retinas RGC density (≈98%). Single IVT dose of
monly used in hospitals; (iii) I.V. route favors the distribution siCASP2 is able to protect RGC cells during 7 days post-
of QPI-1002 to the target renal cells; (iv) only a single admin- ONC. The duration of effects lasted for up to 30 days in
istration of QPI-1002 (10 mg/Kg) is needed, resulting in a animals that received 2 IVT injections of 20 μg siCASP2, at
total amount of ≈ 700 mg for a patient with 70 Kg. If ongoing Days 0 and 10 after ONC. This study also used the model of
phase III gives positive results, the orphan drug status can optic nerve transection (ONT), based on the axotomy of all
speed-up access to the pharmaceutical market. In this sense, RGCs of the left eye. The procedure causes a more aggressive
RNAi-based drugs in phase II – III clinical trials

Fig. 3 QPI-1007 and mRNA target sequences. (a) A schematic representation of QPI-1007 duplex. Passenger (5'→3') and guide (3'→5'). Uppercase letters
represent unmodified nucleotides. Chemical changes are: (i) 2'-O-methyl sugar modified RNA nucleosides, represented by lowercase letters; (ii) L-DNA cytidine
nucleotide, represented by Y; (iii) inverted deoxyabasic moiety, represented by ia. (b) Complementarity between QPI-1007 guide strand and the human and rat
caspase-2 mRNAs NCBI reference sequences, NM_001224.4 and NM_022522.2, respectivelly. Target sequences for QPI-1007 in caspase-2 mRNA are
represented in red (5'→3'), while QPI-1007 guide strands are written in orange (3'→5'). QPI-1007 molecular sequence was previously published.

loss of cells. Animals received 2 doses of siCASP2 (10 μg), the following by a rapid clearance via renal filtration. No
first immediately after ONT and the second at Day 7 post- systemic or genetic toxicity was observed. The QPI-
ONT. SiCASP2 caused a 15% increase in RGC survival 1007-related adverse effect found was only a mild inflam-
compared to the control siRNA at 2 weeks after injury (48). matory cell infiltration in vitreous (49).
Indeed, Vigneswara and Ahmed (2016) recently found that In 2010, Quark started the first clinical trial on QPI-1007
siCASP2 IVT injected every 8 days could protect RGC cells (NCT01064505). A phase 1 clinical study on safety, tolerabil-
for 12 weeks after ONC (50). ity, dose-limiting toxicities, and the pharmacokinetics of a sin-
A preclinical study evaluated the pharmacokinetic and tox- gle IVT injection was executed. Recruited patients presented
icological properties of QPI-1007, to support drug testing in either a chronic optic nerve atrophy - Blegally blind^ in the
human trials (49). Studies were carried on rabbits and rats. study eye (stratum I) or a recent onset of NAION disease, with
Preclinical studies on rabbits evaluated ocular and systemic diagnosis at ≤28 days (stratum II). As a secondary end point,
effects of single or repeated bilateral IVT injections. Doses the study also evaluated if QPI-1007 causes anatomical
and intervals for QPI-1007 IVT injections varied according changes in the optic nerve head and retina or affects visual
to the experimental assays. For pharmacokinetics, single doses acuity and visual field. Quark reported no dose-limiting tox-
of 0.3 mg/eye or 3 mg/eye, and time points from 1 to 336 h icity in 5 of 6 cohorts of patients with chronic optic nerve
post QPI-1007. Single-dose or repeated-doses of 0.3, 1, or atrophy (51). QPI-1007 was well tolerated also in patients with
3 mg/eye were tested for toxicity. In rats, QPI-1007 was ad- NAION. No serious adverse event was found, no dose limiting
ministered by I.V. route via tail vein at single-doses (4, 20, or toxicities and no discontinuation due to adverse effects oc-
100 mg/Kg) or repeated ones (10 or 75 mg/Kg/day, for curred. Patients treated with QPI-1007 (51.7%, n = 29)
28 days). Genotoxicity tests, bacterial mutation assays, chro- showed improvements in visual acuity, as defined by an in-
mosome aberration test, and micronucleus evaluation were crease in BCVA values ≥3 lines (≥ 15 letters) at month 3.
carried on. QPI-1007 presented a half-life (t1/2) of 2 days in Indeed, none of the NAION patients treated with QPI-1007
vitreous and retina/choroid in the rabbit. Repeated IVT in- lost 3 or 2 lines of visual acuity at months 3 and 6, compared to
jections every 2 or 4 weeks for up to 11 doses caused no 9.1% and 14.8% in historical controls. Drug was able to
accumulation. QPI-1007 was chemically stable in vitre- change the natural history of disease (52). The company is
ous, and was eliminated by a slow egress to blood, now recruiting participants for a phase 2/3 clinical trial on
Titze-de-Almeida, David and Titze-de-Almeida

Fig. 4 Schematic representation of a stable-nucleic-acid-lipid particle (SNALP). These particles of < 200 nm in size have provided an efficient delivery system for
siRNAs administered by intravenous route. SNALPs resemble the well-known liposomes, as they are vesicles with an outer lipid bilayer enclosing a water
compartment inside. SNALPs present some valuable improvements for in vivo studies. Negatively-charged siRNA molecules (the small double-stranded
structures, in orange) are incorporated into the aqueous inner part with the help of cationic lipids (1,2-dilinoleyloxy-N,N-dimethyl-3-aminopropane,
DlinDMA). Helper lipids with a yellow head (1,2-distear-oyl-sn-glycero-3-phosphocholine, DSPC) promote the release of siRNAs from SNALPs in cell
cytoplasm. Polyethylene glycosylated lipids (PEG–C–DMA) offers an external surface more hydrophilic and neutral, for stability and crossing barriers.
Cholesterol helps to stabilize the lipid particle formulation. Reprinted with permission from Reference (56). Copyright (2012), Elsevier. DSPC 1,2-distear-oyl-
sn-glycero-3-phosphocholine, PEG–C–DMA Polyethylene glycosylated lipids, PEG–C–DMA Polyethylene glycosylated lipids.

efficacy and safety of QPI-1007 in ≈530 patients with recent- check whether the benefits were statistically significant. From
onset of NAION (NCT02341560). a market point of view, QPI-1007 may be of value for other
Finally, QPI-1007 was tested in subjects with acute attack eye diseases that result in loss of visual acuity and blindness (as
of primary angle-closure glaucoma (APACG) glaucoma, for example), as it prevents apoptosis of retinal
(NCT01965106). This phase 2 study evaluated the safety, cells. Indeed, Quark was granted a patent from the United
tolerability, and pharmacokinetics of a single IVT injection States Patent Trademark Office on the use of QPI-1007 for
of 1.5 mg QPI-1007. The clinical trial also examined the treatment of patients with NAION (53). The protective effects
prevention of damage to nerve tissue in the glaucomatous of this siRNA on retinal cells and consequent ability to im-
eye and changes in visual function.The study finished in prove visual function must be confirmed in a phase 2/3 trial in
July 2015 and no results are available yet. patients with NAION.

Critical Analysis
TKM-080301 (TKM-PLK1)
QPI-1007 followed the same strategy of QPI-1002, as it sup-
presses an apoptotic protein involved in cell death. Compared TKM-080301 targets polo-like kinase 1 (PLK1), a protein
to QPI-1002, QPI-1007 is a siRNA with much more chemical overexpressed in cancer cells promoting an uncontrolled cell
changes for improving its knocking-down action. The target is proliferation (54). Some PLK-1 inhibitors are already in hu-
caspase 2, an enzyme that triggers apoptosis of retinal cells in man clinical trials for anticancer therapy (55). Regarding
ischemic optic neuropathies like NAION. The drug is injected TKM-080301, it is a siRNA for I.V. administration formulat-
by IVT route, like PF-04523655, thus requiring an ophthal- ed in stable nucleic acid lipid particles (SNALPs, as shown in
mologic service. Although QPI-1007 improved visual acuity Fig. 4) (56, 57). The drug is developed by Tekmira (BTKM^
and stabilized the disease progression, the data was compared from siRNA designation TKM-080301), a company that
only with historical controls. A phase 2 controlled study must changed name to Arbutus Biopharma Corp. in 2015.
RNAi-based drugs in phase II – III clinical trials

Preclinical characterization of TKM-080301 included Another clinical trial evaluated safety and antitumor activity
in vitro and in vivo pharmacological activity studies, effects on of TKM-080301 in patients with refractory adrenocortical
immune system and toxicity (58). The drug showed antipro- cancer (ACC). The study used treatment cycles described
liferative activity on cancer cell lines and ability to silence above, and the anticancer activity was evaluated by
PLK-1. TKM-080301 also presented antitumor activity in RECIST 1.1 (Response Evaluation Criteria in Solid Tumors
xenograft models of human cancer. Effects were confirmed 1.1) every two cycles (62). The study enrolled 16 subjects that
by histopathology and molecular methods, showing PLK1 received 0.6 or 0.75 mg/kg/week TKM-080301 for up to
mRNA cleavage products in samples from treated animals. 18 cycles. Four out of eight patients that received at least
A single dose of TKM-080301 caused PLK1 silencing that two cycles of treatment showed an improvement of stable
persisted for 7–10 days. No measurable immune activation disease. One of them presented a 13% decrease in tumor
was found. Toxicity was restricted to liver and spleen, without diameter. Indeed, a metastatic intraperitoneal non-
myelosuppressive effects. Preclinical data highlighted PLK1 as functional ACC showed a tumor reduction of 19% after cycle
a promising anticancer target and supported the clinical eval- 2 and 49% after cycle 14. Some serious adverse effects caused
uation of TKM-080301. by TKM-080301 were ECG T-wave inversion, musculoskel-
The first phase 1/2 clinical trial of TKM-080301 etal pain and infusion reaction. Despite adverse reactions, this
(NCT02191878) evaluated safety and toxicity. Primary out- study revealed that: 1- TKM-080301 was generally well tol-
come measures were the maximum tolerated dose (MTD) and erated at 0.6–0.75 mg/Kg for up to 18 cycles and; 2- the
dose-limiting toxicities (DLT), as determined in a dose escala- siRNA presented an antitumor activity in patients with refrac-
tion study. Sequential cohorts of 3 to 6 patients, with either tory ACC (62).
advanced solid tumors (i.e. advanced hepatocellular carcino-
ma - HCC) or lymphomas, received escalating doses of TKM- Critical Analysis
080301 (0.15 to 0.90 mg/Kg) in cycles of 28 days of treatment
(59). In each cycle, the siRNA was administered as a 30-min Proposed regimen for TKM-080301 (18 cycles X 3 injections
IV infusion once a week for three consecutive weeks (at days 1, per cycle X 0.75 mg/Kg) results in a high amount of siRNA of
8, and 15) followed by a 1-week rest period. Common adverse ≈ 2800 mg, but it shows antitumor activity in about 50% of
effects were rigors (33%) and fever (25%) (60, 61). Mild to patients with advanced hepatocellular carcinoma or refracto-
moderate delayed infusion-related reactions were observed. ry adrenocortical cancer. As PLK1 is deregulated in various
Some cytokines were upregulated, like interleukins (IL) IL-6, types of cancer, this siRNA may have an increased market in
IL-8, and monocyte chemoattractant protein-1 (MCP-1) at the future. However, PLK1 is a druggable target. PLK inhib-
doses ≤0.75 mg/Kg/week (59). DLT occurred in two patients itors already in clinical trials (like volasertib in phase 3) would
at 0.9 mg/Kg/week with adverse grade 3 reactions. Thus be potential competitors of TKM-080301 (63). Regarding
MTD was defined as 0.75 mg/Kg/week, and this dose was adverse effects, patients showed mild to moderate infusion-
adopted for subsequent studies. The authors concluded the related reactions with cytokines upregulated - a common con-
siRNA was generally well-tolerated. Secondary outcomes in- sequence of lipid-carried siRNAs administered by I.V. route.
cluded evaluation of pharmacokinetic parameters and a pre- The cost of therapy could be acceptable if phase 2 and 3
liminary antitumor activity. Pharmacokinetic studies in cycles confirms impressive preliminary results.
1 and 2 revealed a dose proportional Cmax (maximum plasma
concentration) and AUC (area under the curve) and no accu- ATU027
mulation. Even 6–8 cycles of TKM-080301 has caused no
cumulative toxicity in two individuals. TKM-080301 present- Atu027 is a chemically synthesized siRNA designed to silence
ed a preliminary antitumor efficacy (59). Clinical benefits were protein kinase N3 (PKN3) mRNA. PKN3 is a downstream
found in 4 of 9 (44%) patients treated with ≥0.6 mg/Kg effector of the phosphoinositide 3-kinase (PI3) signaling cas-
TKM-080301 (61). An expansion cohort (n = 10 patients) cade in highly vascularized tissues (64, 65). Acting as an
was enrolled in 2013 aimed to explore pharmacodynamic Bendothelium-modulating^ drug, it stabilizes vessel integrity,
data and antitumor activity from biopsy samples at previously counteracts inflammation in tumor region, and reduces the
determined phase 2 dose, the MTD of 0.75 mg/Kg/week spread of metastatic cells (66, 67). The target is the PKN3-
[(61); NCT01262235]. Authors compared biopsy specimens mRNA sequence 5′-agacuugaggacuuccuggacaa-3′ (65, 68). It
(for example, same lesion of the right lobe liver) pre- and two is a double-stranded 23-mer nucleotide, blunt-ended, and
days post-TKM-080301 dose 1. They found a 476-bp PLK1 chemically stabilized by alternating 2′-O-methyl-nucleotides.
mRNA cleavage product after treatment, by using 5′-RACE Atu027 is formulated in lipid particles (siRNA-lipoplex,
(rapid amplification of cDNA ends) polymerase chain reaction Atuplex®) for systemic administration and delivers PKN3-
(PCR). Predicted cleavage PLK-1 amplicons were observed in targeted siRNAs into the cytoplasm of vascular endothelial
agarose gel electrophoresis and confirmed by sequencing (61). cells in vivo. The lipoplex contains positively charged liposomes
Titze-de-Almeida, David and Titze-de-Almeida

to which the negatively charged siRNAs are attracted and Another study with Atu027 was a phase 1/2 clinical trial
form complexes (69). The four components of Atu027 are (NCT01808638). This work evaluated whether Atu027 could
the following: 1- PKN3-targeted siRNA described above; 2- be administered in association with the standard chemother-
cationic lipid AtuFECT01; 3- neutral helper lipid; 4- a apeutic gemcitabine for patients with advanced or metastatic
pegylated lipid (65, 69). pancreatic adenocarcinoma. The clinical trial evaluated two
Preclinical studies examined the antitumor efficacy of dose regimens of Atu027. In arm 1, the 28-day cycle was
Atu027. This siRNA was intravenously administered to mice, composed by weekly injections of Atu027 and gemcitabine
rats and cynomolgus monkeys, causing an RNAi-mediated for three consecutive weeks (days 1, 8, and 15) followed by a
knock-down of PKN3. Regarding anticancer effects, it re- week with no treatment. Patients received a total of six admin-
duced tumor growth and lymph node metastasis in orthotopic istrations of this siRNA in 8 weeks. Cycles were repeated,
mouse models (65). The authors found a significant reduction except if toxicity or disease progression occurred. Patients en-
in lymph vessel density, but not in microvascular density. rolled in arm 2 also received gemcitabine once weekly (days 4,
Atu027 also prevented the formation of lung metastasis, as 11, and 18), but Atu027 was given twice weekly (days 1, 4, 8,
found in two experimental mice models (66). 11, 15, 18, 22, and 25). Thus they received eight doses (33%
The first-in-human phase 1 clinical trial evaluated the safe- more Atu027 than patients of study arm 1). In this arm 2, the
ty, tolerability, pharmacokinetics, pharmacodynamics, and combination cycle was followed by a 28-day treatment with
anticancer activity of Atu027 on single and repeated infusions only gemcitabine. The company reported Atu027 was well
in patients with advanced refractory solid tumors tolerated in combination with gemcitabine for patients with
(NCT00938574) (67). Patients first received the drug as a sin- pancreatic cancer. A higher dose of Atu027 tested in study
gle treatment, followed by a 3-week safety period for clinical Arm 2 provided a longer duration of progression-free survival
examinations. Then eight infusions twice per week over a 28- (PFS) rate (5.33 months versus 1.81). The difference was statis-
day cycle were administered. Each lyophilized Atu027 dose tically significant for patients with metastatic pancreatic can-
was diluted in 5% xylitol, and was administered via I.V. route cer. Overall survival (OS) was also higher in patients of study
over four hours. Cohorts of patients (n = 3 to 7) were exam- arm 2 in comparison with those of arm 1 (70).
ined for drug safety before dose escalation, i.e., three patients
per dose level extended by at least another three for definition Critical Analysis
of MTD (the B3 + 3^ design scheme). The study enrolled
thirty-four patients with advanced refractory cancer for testing Treatment of pancreatic cancer remains a great challenge in
10 escalating doses of Atu027 (0.001 – 0.336 mg/kg) (67). oncology (71). Atu027 is also a formulated anticancer siRNA,
Regarding pharmacokinetics, I.V. administration of Atu027 with anti-angiogenic and anti-metastatic properties. It re-
caused a rapid increase in siRNA plasma concentration dur- quires a smaller dose (0.336 mg/Kg) and fewer injections
ing the 4 h infusion, followed by a steep decrease. Maximal (n = 8) than TKM-080301, resulting in a lower total dose of
siRNA level (Cmax) was 180 ng/mL, achieved for Atu027 dose ≈ 190 mg (8 injections X 0.336 mg/Kg X 70 Kg). I.V. injec-
of 0.336 mg/Kg at 2 h post injection (Tmax). Half-life values tion increased C3 proteins that indicate an immune activa-
for single and eighth repeated siRNA injections were 16 h and tion. Pancreatic cancer is a very aggressive and metastatic
10 h, respectively. Regarding toxicity, MTD was not achieved disease (72). Thus Atu027 was associated with gemcitabine,
as only one patient showed a grade 3 adverse reaction possible which improved the progression-free survival (PFS) rate in
related to Atu027 at the highest dose (0.336 mg/Kg). This patients with advanced- and metastatic pancreatic adenocar-
siRNA was well-tolerated; less than 10% adverse events were cinoma. This siRNA is a promising alternative for patients
grade 3 or greater, and most of them resulted in complete with incurable pancreatic carcinoma to be examined in a
recovery. They were not related to escalating dosage. About further phase 3 clinical study.
immune response commonly found for particulate com-
pounds, this study showed a dose-dependent increase in C3 SYL040012 (Bamosiran)
protein that is a marker of complement activation. No clini-
cally significant changes were found for interferon alfa, IL-1β, SYL040012 is a siRNA targeting β2-adrenergic receptor, de-
IL-6, tumor necrosis factor α, and MCP-1. Also, no antibodies signed for glaucoma treatment (2). Glaucoma is an ophthal-
against siRNA Atu027 molecule were detected. Finally, mic disease that causes visual impairment, leading to blindness
Atu027 was effective in 52% patients (14 of 27). They showed as a result of progressive optic nerve damage (73). The main
disease stabilization after repeated treatment (week 8), ac- risk factors for glaucoma are elevated intraocular pressure
cording to RECIST criteria. Eight of them still had a (IOP), genetic and ethnic backgrounds, and age (74). IOP is
stable disease at week 12. Some patients also showed caused by an unbalance between production and drainage of
less metastatic lesions (i.e. lung, lymph node, liver, bone) the aqueous humor (AH) (75). Drugs that reduce AH produc-
after Atu027 treatment (67). tion and control IOP are valuable for patients with glaucoma.
RNAi-based drugs in phase II – III clinical trials

β-blockers produce this effect and can be prescribed for this first study interval, Interval 1 (n = 6 individuals), SYL040012
disease (76). Instead of blocking β-receptors proteins, another was instilled in a single dose of 600 μg/eye/day. Only after the
strategy is to knock-down the corresponding mRNA by RNAi. tolerability criterion (i.e. absence of grade 3 or higher toxicity)
SYL040012 is a naked 21-base synthetic siRNA targeting the was meet at 72 h, the next individual was tested. In the
β2-adrenergic receptor mRNA. Interval 2, the drug was given in daily instillations over seven
In preclinical testing, SYL040012 (100 nmol/l) reduced consecutive days. Two doses tested were 600 and 900 μg/
50% of β2 receptor mRNA content in cells in culture at eye/day (n = 12 individuals each dose). Ophthalmic tolerance,
24 h – 48 h after transfection (2). siRNA showed no significant adverse effects, vital signs (cardiovascular and respiratory pa-
effects on other adrenergic receptors, β1 or α1B, revealing its rameters), clinical biochemistry, pharmacokinetic from blood
strict specificity. This naked siRNA rapidly enters the eye and samples, and IOP at various time points post instillation were
reaches various structures, including producing HA like ciliary measured. As a result, no ocular pathological changes were
body. Fortunately, only low levels of SYL040012 duplex (not observed at any time. Single and repeated administrations of
biologically active) reached the plasma and systemic organs. SYL040012 were safe and well tolerated. Six mild adverse
The ocular route is advantageous for administering siRNAs; events were observed, and half of them considered unrelated
the first RNAi drugs entering clinical trials attended ocular to the drug. SYL040012 was not detected in blood samples
diseases (77–79). Eyes are anatomical structures placed exter- after a single or repeated administration. For IOP, the repeat-
nal and apart from the body. Administrating drugs topically ed dose schedule of Interval 2 reduced IOP in 15 of 24 healthy
by eye drop solutions reduces their distribution to the systemic subjects. After four days of administration of 600 μg/eye/day,
circulation. The amount of drug administered is lower com- SYL040012 caused an overall statistically significant reduc-
pared to other routes of administration. Finally, the immune tion in IOP. The effect was more prominent in individuals
responses are relatively lower in the eyes, reducing the risk of (n = 5) with higher basal IOP values.
undesired reactions against therapeutic nucleotides (80). Three other clinical trials were also completed, as follows:
The study cited above also measured the efficacy of (i) Phase 1/2 evaluated tolerance and drug efficacy in unmed-
SYL040012 for IOP reduction in New Zeland White icated patients with elevated IOP (NCT01227291). (ii) Phase
Rabbits (2). The authors first tested the model with two drugs 2 study of tolerability and effects of SYL040012 on IOP of
used for glaucoma treatment, dorzolamide and latanoprost. patients with ocular hypertension or open-angle glaucoma
Both successfully reduced IOP for six hours. These drugs, (NCT01739244). SYL040012 was instilled daily over 14 days
however, are not β-blockers, limiting the informative val- to 89 patients, organized in groups receiving the following
ue of this test. Rabbits present β2-receptors widely distrib- doses: 80 μg/eye/day (0.2%), 300 μg/eye/day (0.75%),
uted in ocular tissues, mainly in conjunctival, corneal, and 900 μg/eye/day (2.25%), or placebo (83). The drug was well
ciliary process (81). tolerated. Adverse reactions occurred in 14.6% of the pa-
In SYL040012 preclinical testing, a topical dose of tients, and most of them were mild. SYL040012 caused a
20 nmol/day for four consecutive days caused ≈ 22% significant reduction in IOP at the dose 300 μg/eye/day, in
decrease in IOP in healthy animals. The drug also showed comparison with placebo group or the basal values (i.e. IOP
efficacy in a glaucoma rabbit model named Boral water curve obtained in screening period). (iii) Phase 2 clinical trial
overloading^. Animals first received 40 nmol/eye/day also named SYLTAG evaluated four doses of SYL040012 eye
for four consecutive days, and then ocular hypertension drops, in comparison with timolol maleate 0.5%, for lowering
was induced by water loading. SYL040012 caused a sig- IOP in patients with open-angle glaucoma (NCT02250612)
nificant reduction in IOP compared to the PBS-control (84). Effects of each siRNA concentration on IOP and the
group. As expected, the content of β2 receptor mRNA local and systemic safety were evaluated after a 28-day treat-
was reduced in lacrimal glands and ciliary body at 24 h ment. This multicenter study enrolled 184 patients, that were
after the last SYL040012 dose. Safety was tested in randomized into groups receiving 150 – 600 μg of
cynomolgus monkeys. They received topical ocular instilla- SYL040012 daily and 0.5% timolol twice a day. All treat-
tion of SYL040012 (72, 144, and 288 nmol/eye/day) dur- ments reduced IOP at Day 28 with the highest effect - com-
ing a period of 28 days. Clinical and histopathological pared to the baseline – occurring for SYL040012 300 μg. In
examination revealed the drug was safe and well tolerated the total population, SYL040012 had not achieved the objec-
(2). tive of non-inferiority compared to timolol (85). However, in a
The first human phase 1 clinical trial on SYL040012 subgroup of patients with IOP >25 mmHg, SYL040012
(NCT00990743) evaluated the safety, tolerability, and bio- 450 μg was non-inferior to timolol. Regarding toxicity, no
availability in thirty healthy volunteers with IOP below grade 3 or 4 adverse reactions occurred in the 28-day treat-
21 mmHg (82). The effect of different siRNA doses on IOP ment period. Furthermore, adverse events were less frequent
was also followed. The drug was instilled as eye drops in one in all SYL040012 groups (8% - 12%) than in timolol group
eye, and the untreated fellow eye served as a control. In the (24%) (84).
Titze-de-Almeida, David and Titze-de-Almeida

Critical Analysis Zealand white rabbits. SYL1001 showed efficacy for ocular
irritation, reducing TRPV levels and ocular pain caused by
SYL040012 addresses glaucoma, a disease currently treated capsaicin. Analgesia was comparable to that obtained by the
with eye drops containing β-receptor antagonists, prostaglan- reference analgesic capsazepine. For toxicological analyses,
dins, and others. Prostaglandin analogs have been preferen- two different species - dogs and rabbits - received ocular in-
tially prescribed compared to β-blockers due to the safety pro- stillations of SYL1001 during 28 days. No toxic effect was
file and patient compliance (76). The rationale of SYL040012 found. Regarding pharmacokinetics, the drug was not detect-
as a selective β2 suppressive drug is interesting regarding safety ed above the limit of detection of 40 ng/ml in all time points
since non-selective blockers like timolol are relatively contra- and doses administered. In summary, SYL1001 presented
indicated for patients with cardiac problems (86). There is a safety and efficacy in the model of ocular pain induced by
risk of systemic absorption via the nasolacrimal system and capsaicin (90).
following side effects from blockade of β1 heart receptors A pharmacokinetic study evaluated the biodistribution of
(87). However, suppression of β2 receptors by SYL0400012 SYL1001 in New Zealand rabbits (91). Cornea, conjunctiva,
might cause bronchoconstriction in patients with asthma and and lachrymal gland were harvested at 5 min after eye drop
chronic obstructive pulmonary diseases (88). Fortunately, the instillation. Also, the assay collected trigeminal/semilunar
phase 1 study did not detect the drug in blood samples, prob- ganglion to examine whether SYL1001 had access to neurons
ably due to the rapid attack of plasmatic ribonucleases on a innervating the cornea. To disclose systemic exposure, sam-
siRNA molecule that was not protected either by chemical ples of lung, liver, kidney, and plasma were analyzed.
changes (i.e. phosphorothioate linkages) or a nanoparticle. In SYL1001 was detected in all ocular structures and trigeminal
fact, SYL0400012 caused less adverse events than timolol. ganglion, and also in plasma. Regarding human clinical trials,
This siRNA showed activity in comparison with placebo. a phase 1 study evaluated safety and tolerability of this siRNA
However, it was not equivalent to timolol except in patients (NCT01438281). Thirty healthy individuals received
with high IOP, which reduces the potential market for a pro- SYL1001 by eye drops. For an initial evaluation of safety,
spective drug. The total amount of 13 mg (28 daily doses X the drug was administered to six subjects in a single dose.
450 μg per dose) is relatively high for an eye drops siRNA. Then escalating doses were given to 24 individuals, as a single
Thus the cost-benefit relation is problematic. From a general daily dose, in one eye, for seven days. The drug was well
view, SYL0400012 was the first siRNA tested in humans for tolerated, without causing changes in ocular surface or severe
topical use in eyes, bringing a contribution for the biotechnol- adverse effects. Regarding pharmacokinetic analysis,
ogy of RNAi. At that time, chemical changes in siRNA mol- SYL1001 was not detected in plasma above the limit of quan-
ecules that increase both duration of activity tification of 40 ng/ml (92).
(phosphorothioated backbones that avoid ribonuclease attack) Two phase 2 clinical trials evaluated safety and efficacy of
and target specificity (LNA, locked-nuclei acids) were not yet SYL1001 in patients with ocular pain associated with Dry Eye
available. Even nanocarriers for in vivo use were in their infan- Syndrome (NCT01776658 and NCT02455999). Analgesic
cy. The question is how much this biotechnological platform effect and ocular tolerance of four doses of SYL1001
could improve in terms of efficacy and dosing regimen wheth- (0.375%, 0.75%, 1.125% and 2.25%), including a placebo
er it received new layers of technology, like: (i) chemical group, were evaluated after ten days of treatment of 127 pa-
changes in siRNA structure; (ii) nanoparticles delivery; (iii) tients (93). The drug caused analgesia in Visual Analogue
siRNA structural changes to single-stranded antisense oligo- Scale (VAS), which was significantly better than placebo
nucleotide, like a GapmeR for example, that can be formu- for dose 1.125%. The analgesic effect measured by
lated in particles or administered naked. Ocular Surface Disease Index (OSDI) scale, however,
showed no difference between treated and control groups.
SYL1001 No serious adverse events occurred, either locally in the
eye or systemically (93).
SYL1001 is a siRNA designed for prevention and treatment of
ocular pain and Dry Eye Syndrome, formulated for topical Critical Analysis
administration by eye drops. The target of SYL1001 is the
vanilloid receptor TRPV1 (transient receptor potential cation The treatment of Dry Eye Disease is still a challenge in oph-
channel, subfamily V, member 1), a nociceptor found in cor- thalmology, including cyclosporine, anti-inflammatory drugs,
nea and its respective nerve terminals (89). artificial tears, and other drugs (94, 95). SYL1001 does not act
Preclinical testing of SYL1001 used the rabbit model of in the disease pathogenesis. But it is an attractive analgesic for
ocular irritation induced by the capsaicin, a natural agonist the ocular pain faced by patients, as it blocks the neuronal
of TRPV1 (90). This study first validated different TRPV1- signaling at its origin in TRPV1 nociceptors. Administered
targeted siRNAs by in vitro assays and tested them in New dose of SYL1001 is moderate, making possible an affordable
RNAi-based drugs in phase II – III clinical trials

Fig. 5 Rationale on Atu027 technology for the treatment of TTR-mediated amyloidosis. (a) Patients with amyloidosis present an increased expression of TTR
gene. Atu027 is a TTR-targeted siRNA, formulated in lipid carriers for intravenous administration. (b) Lipid particles coated with ApoE bind to receptors in hepatic
cells and are internalized by endocytosis. (c) Ionizable lipids of these particles will acquire positive charges by protonation due to the acid pH of endocytic vesicles.
Positively charged nanoparticles will bind to negatively charged endosomal membrane, and the fusion of these membranes will finally deliver siRNAs in cell
cytoplasm. (d) TTR-targeted siRNAs will bind to complementary sequences in 3' end of TTR mRNA, instead of the traditional CDS region. AGO enzyme of RISC
complex will execute TTR mRNA cleavage. (e) Reducing TTR expression will prevent the accumulation of abnormal amyloid proteins in tissues. AGO Argonaute
2, ApoE apolipoprotein E, CDS Coding sequence region of mRNA, Dicer a ribonuclease enzyme PACT Protein activator of the interferon-induced protein kinase
(PKR), RISC RNA-induced silencing complex, TRBP HIV-1 Trans-activation response (TAR) RNA-binding protein, TTR transthyretin.

treatment. At 450 μg/eye/day, it causes analgesia, and a ten- mediated amyloidosis (ATTR), characterized by an accumu-
day regimen will demand 4.5 mg of siRNA per eye. Other lation of abnormal insoluble amyloid proteins in tissues, par-
cycles of treatment might be necessary for these chronic eye ticularly peripheral nerves and heart (96). Prognosis for pa-
disorder. A phase 2 study with ≈ 130 individuals showed a tients with TTR is poor. Current treatments are aimed to
significant analgesic effect in one of two tests of analgesia at reduce mutant TTR or to stabilize the correct conformation
450 μg per eye (1.125%). As pain is a clinical symptom present of TTR protein, but these drugs are not effective in all patients
in other prevalent eye diseases, the pharmaceutical market of (97). The therapeutic focus of patisiran is the familial
SYL1001 is significant. Regarding Dry Eye Disease, it is a amyloidotic polyneuropathy (FAP) related to TTR mutations.
complex disorder for therapeutic management. The contribu- This neurological disease is progressive, debilitating, and irre-
tion of SYL1001 would be only for analgesia, not replacing versible, with symptoms of sensorimotor and autonomic neu-
the immunosuppressive agent cyclosporine and other drugs ropathy (98). Patisiran is a potent antitransthyretin siRNA
for symptomatic relief. For that, a multicenter phase III study encapsulated in lipid nanoparticles, which targets a conserved
is needed to examine whether this siRNA will show efficiency sequence present in TTR mRNA. Curiously, this sequence is
and safety in a large number of individuals. located in noncoding 3′-UTR region of TTR mRNA. siRNAs
usually target coding regions of mRNA while microRNAs
Patisiran (ALN-TTR02) address noncoding 3′-UTR ones (79). The author’s argument
is that the same siRNA target sequence was present in both
Patisiran is a siRNA that targets transthyretin (TTR) mRNA. the nonmutated and mutated TTR mRNA, resulting in a
Mutations in TTR gene cause a disease named transthyretin- more efficient gene knocking-down (99). Furthermore, the
Titze-de-Almeida, David and Titze-de-Almeida

siRNA molecules of patisiran were formulated in a lipid car- Twenty-nine adult patients with biopsy-proven ATTR am-
rier previously developed by Tekmira. The benefit of this yloidosis and mild-to-moderate neuropathy were organized
carrier is that it vectorizes patisiran to the liver, where most in cohorts of individuals. They received two I.V. infusions
TTR proteins are synthesized. Lipid particles are coated with of patisiran, at the following dose regimens: 0.01 mg/Kg
the apolipoprotein E (ApoE). After I.V. injection, the carrier is (n = 4), 0.05 mg/Kg (n = 3), 0.15 mg/Kg (n = 3), or
opsonized by this protein, enter the liver and bind to ApoE 0.3 mg/Kg (n = 7) every 4 weeks (Q4W), or 0.3 mg/kg
receptors on hepatocytes. Within the hepatic cells, patisiran’s (n = 12) every 3 weeks (Q3W). To prevent infusion-related
siRNAs will knock-down TTR expression by the standard reactions, patients were premedicated with dexamethasone,
post-transcriptional mechanism, using RISC complex and paracetamol, and antagonists of H1 and H2 histaminergic
Argonaute protein (Fig. 5). receptors. Most individuals presented Val30Met TTR mu-
Before entering phase 1 human clinical trials, patisiran tation, the most common form of FAP (102), and were
was evaluated in cynomolgus monkeys (99). TTR target taking concomitant treatments with tafamidis or diflunisal.
sequence is identical in this species and humans, allowing Regarding pharmacodynamic, patisiran 0.3 mg/Kg Q3W
patisiran testing without nucleotide changes in its siRNA caused ≈ 83–87% TTR knock down after the first and
molecule. Patisiran caused a marked 75% - 90% TTR second doses, respectively, in comparison to baseline levels.
knockdown in monkeys at Day 7 (0.1 mg/Kg) and Day Similar results occurred for Q4W regimen at the same
14 (0.3 mg/Kg), respectively. The silencing effect persisted dose. Patisiran caused the same effect on wild-type and
for 28 days. The drug also showed efficacy in a multiple mutant TTR and showed efficacy also for patients treated
dose regimen - 0.3 mg/Kg every four weeks for five with tafamidis and diflunisal who presented an increased
doses. baseline serum levels of TTR. Mean values of Cmax and
The first human trial on patisiran was a dose-escalating AUC for the first and second doses increased proportion-
study that evaluated safety and efficacy in 17 healthy adult ally to patisiran dose increment. Terminal half-life values
volunteers (NCT01559077) (99). Subjects organized in were 39–59 h for doses over 0.01 mg/Kg, at Day 0 and
groups of four individuals received I.V. injected siRNAs Days 21/28. Regarding safety and tolerability, only two
(0.01–0.5 mg per kilogram). Individuals also received the patients treated with 0.3 mg/Kg showed serious adverse
following drugs to reduce the risk of infusion-related reac- events. Mild-to-moderate infusion-related reactions oc-
tions: dexamethasone, acetaminophen, diphenhydramine curred in 10.3% patients that received 0.3 mg/Kg
or cetirizine, and ranitidine, as previously cited (100). Q4W. In summary, patisiran was generally well tolerated
Patisiran activity was revealed by a decrease in serum and caused a significant knockdown of mutant and wild-
transthyretin levels, measured by enzyme-linked immuno- type TTR in patients with FAP (101).
sorbent assay (ELISA) (99). Doses ≥0.15 mg/Kg caused a The same patients of the above-described phase 2 clinical
significant and durable TTR knock down until Day 28. trial (NCT01617967) were enrolled in phase 2 open-label
The highest siRNA dose (0.5 mg/Kg) caused the best extension study (phase 2 OLE study) (NCT01961921), that
TTR silencing effect (93.8%). 5′ RACE assay confirmed evaluated the safety and tolerability of patisiran 0.3 mg/Kg
the specificity and RNAi-based mechanism of action of Q3W for long periods (≈ seven months). Patisiran was well
patisiran, generating TTR mRNA cleavage products. For tolerated, with ≈ 15% of infusion-related reactions. No sys-
pharmacokinetics, siRNA levels were measured by ELISA- temic toxicity was observed. This siRNA caused a marked
based hybridization assay. Cmax and AUC increased in a 80% TTR knockdown over nine months. Regarding thera-
dose-proportional manner. Finally, the study monitored peutic effects, patients were evaluated by using a modified
adverse effects during the 28-day period after drug admin- Neuropathy Impairment Score + 7 (mNIS + 7), and a quality
istration. No serious adverse effects occurred. The overall of life (QOL) measurement. Six months of patisiran stabilized
incidence of adverse reactions was 7.7%. A moderate the neurological impairment and QOL in patients with FAP,
infusion-related reaction was observed in a single individ- independent on other concomitant treatments (103). This on-
ual who received 0.5 mg/Kg, which was managed by a going study also showed positive effects on mNIS + 7 at
temporary interruption in patisiran infusion, glucocorticoid 24 months of treatment, with only mild infusion-related ad-
therapy, and reducing the rate of infusion. Another phase verse effects (104).
1 clinical trial addressed safety, tolerability, and PK of Finally, patisiran entered in a phase 3 trial named
patisiran in Japanese healthy subjects (NCT02053454). APOLLO study (NCT01960348), aimed to evaluate its safety
Phase 1 results above described (NCT01559077) and efficacy in patients with FAP. Another phase 3 trial is now
allowed patisiran to enter in phase 2 clinical trial enrolling participants who have completed a prior patisiran
(NCT01617967). This study evaluated the safety, tolerabil- clinical study (NCT02510261). The aim is to evaluate safety
ity, pharmacokinetics, and pharmacodynamic of multiple and efficacy of patisiran in patients with FAP in a long-term
escalating doses of patisiran in patients with FAP (101). dosing, i.e. 52 weeks.
RNAi-based drugs in phase II – III clinical trials

Fig. 6 Characterization of sig12D-LODER devices and injection procedure (a) Schematic representation of the siG12D-LODER matrix of PLGA. (b) The
picture is showing the millimetric dimension of sig12D-LODER devices. (c) Standard procedure EUS guidance for implanting devices into pancreatic tumor. (d)
Sustained release of siRNAs (10 μg) from the PLGA matrix over two months. Devices were incubated in PBS (pH = 7.4), at 37 °C. Reprinted with permission
from References (109, 112). KRAS Kirsten rat sarcoma viral oncogene homolog, KRAS-target siRNA siRNA designed to silence mutated KRAS, PLGA matrix
polymeric matrix of poly(lactic-co-glycolic) acid.

Critical Analysis efficacy of patisiran for patients with ATTR, a genetic disease
that urgently need new therapeutic options.
Patisiran is an excellent example of Gene Therapy. This strat-
egy to treat genetic diseases brought exciting discussions for siG12D-LODER
scientific community of the early seventies (105). The method
for gene therapy used by patisiran would be available only two siG12D-LODER is a polymeric system complex designed for
decades later (106). This formulated siRNA was designed for a sustained delivery of a G12D-mutated KRAS-targeted
ATTR, a genetic disease with a major medical need based on siRNA, developed to treat human pancreatic cancer (109,
its significant morbidity and mortality (107). Liver transplan- 110). As shown in Fig. 6a, b, LOcal Drug EluteR (LODER)
tation and treatment with BTTR stabilizers^ tafamidis or is a polymeric matrix of poly(lactic-co-glycolic) acid (PLGA) in
diflunisal are therapeutic options (108). Instead, patisiran trig- a shape of a small cylindrical rod of diameter 0.8 mm and
gers RNAi on TTR gene, thus controlling ATTR at a very length 5.5 mm ± 1 mm.
fundamental step of disease pathogenesis - the mRNA of the Anticancer effects of this anti-KRAS drug and the ability of
gene that causes amyloidosis. TTR knocking down reaches LODER for long term delivery of siRNAs were evaluated
80%. Treated patients showed stabilization of illness progres- (109). First, the cylindrical shape remained unchanged after
sion and improvement in the quality of life. Part of the success 45 days of implantation of siG12D-LODER. This molecular
relies on the carrier that executes a site-specific delivery of device delivers siG12D siRNAs over two months when incu-
patisiran into hepatic tissues. However, hospital assistance bated in phosphate buffer solution (Fig. 6d), and protects from
for I.V. injections and drugs for controlling infusion-related degradation in mouse liver tissue ex-vivo. Pancreatic carcinoma
reactions are additional costs to be considered. The basic drug cells exposed to siG12D-LODER showed reduced KRAS
costs could be an issue as treatment is expected to be for long mRNA and protein level, along with a reduced cancer cell
term, reaching ≈ 670 mg (0.3 mg/Kg, every three weeks for viability. Cells also showed a reduction in epithelial-
24 months). Patisiran is the most advanced siRNA drug in mesenchymal transition, represented by lower cell migration
development, with encouraging results in phase II trial. Two and contact inhibition. SiG12-LODER silenced KRAS expres-
ongoing phase III studies will probably confirm the safety and sion in subcutaneous tumors in vivo, with no toxicity in male or
Table I Synthetic RNAi-Based Drugs In Phase 2–3 Clinical Trials

Drug Target (cell role) Chemistry / formulation Route Disease Phase Status/completion Company/ Refs
collaborator

PF-04523655 RTP801 / Naked siRNA, O-methylated IVT AMD, DME II Completed/2013 Quark / Pfizer 16, 18
(PF-655) (hypoxia-inducible)
QPI-1002*1 p53 (apoptotic) Naked siRNA, O-methylated IV AKI II Recruiting / 2018*2 Quark 40
(I5NP) DGF III Recruiting / 2019*2
QPI-1007 CASP2 (apoptotic) Naked siRNA, O-methylated; IVT NAION II / III Recruiting / 2019*2 Quark 52
changes in sense strand Glaucoma II Completed / 2015
TKM-080301 PLK1 (kinase) siRNA / SNALP IV Solid tumors, HCC, NET, I / II Completed / 2015 Arbutus 59–62
(TKM-PLK1) ACC, lymphoma
Atu027 PKN3 (kinase) siRNA / LIPOPLEX IV Pancreatic cancer I / II Completed / 2016 Silence / Granzer, 67, 70
FGK
SYL040012 ADRB2 (β2 receptor) Naked siRNA Eye drops Ocular hypertension, glaucoma II Completed / 2013; 2016 Sylentis 82–85
(Bamosiran)
SYL1001 TRPV1 (nociceptor) Naked siRNA Eye drops Ocular pain in Dry Eye II Completed / 2016 Sylentis 92, 93
Syndrome
Patisiran TTR (amyloidogenic) siRNA / Lipid particle, ApoE IV TTR-mediated Amyloidosis III Active / 2017*2 Alnylam 99, 101, 103,
(ALN-TTR02) 104
siG12D-LODER KRAS (oncogene, siRNA / Miniature PLGA Intra- Pancreatic cancer*3 II Active, not yet recruiting / Silenseed 112
GTPase) device tumoral 2020*2
Miravirsen miR-122 (microRNA) AntimiR, antisense SC Hepatitis C infection II Complete / 2011 Santaris 119, 126–128
oligodeoxynucleotide,
LNA, PS

ACC adrenocortical carcinoma; ADRB2 adrenoceptor beta 2; AKI acute kidney injury after cardiac surgery; AMD Age-Related Macular Degeneration; ApoE apolipoprotein E; CASP2 Caspase 2; DGF Delayed Graft
Function in kidney transplantation; DME Diabetic Macular Edema; HCC hepatocellular carcinoma; IV intravenous injection; IVT intravitreal injection; KRAS Kirsten rat sarcoma viral oncogene homolog; LNA locked
nucleic acid; miR-122 microRNA 122; NAION Acute Non-Arteritic Anterior Ischemic Optic Neuropathy; NET neuroendocrine tumors; PKN3 protein kinase N3; PLK1 Polo-like kinase-1; PS phosphorothioated; SC
subcutaneous injection; SNALP stable-nucleic-acid-lipid particles; TRPV1 transient receptor potential vanilloid 1; TTR transthyretin. *1- drug licensed to Novartis; *2- estimated completion data; *3- siG12D-LODERs
combined with chemotherapy treatment (Gemcitabine + nab-Paclitaxel). Company names: Quark Pharmaceuticals; Pfizer; Arbutus Biopharma Corporation; Silence Therapeutics GmbH; Granzer Regulatory
Consulting and Services; FGK Clinical Research GmbH; Sylentis, S.A.; Alnylam Pharmaceuticals; Silenseed Ltd.; Santaris Pharma A/S
Titze-de-Almeida, David and Titze-de-Almeida
RNAi-based drugs in phase II – III clinical trials

female mice or rats, at up to 320 μg siG12D/Kg body weight. unresectable LAPC will receive a single dose of 2.8 mg (i.e.
At this dose, it reduced tumor growth in vivo. Rats showed larger eight 0.35 mg siGD-LODERs) by EUS, in combination with
necrotic areas in tumor region and mice presented smaller chemotherapy (gemcitabine + nab-paclitaxel).
tumor volumes. Survival period significantly increased in
siG12-LODER treated mice. The treatment also showed ben- Critical Analysis
efits in orthotopic pancreatic tumors in mice. Overall survival
was prolonged while KRAS expression was reduced. At first, siG12D-LODER made KRAS a druggable antican-
The last preclinical studies for safety, toxicity, and cer target. New drugs also targeting this oncogene are now in
toxicokinetics were performed in Sprague-Dawley rats. test an may represent future competitors of siG12D-LODER
This anti-KRAS RNAi system was implanted in subcu- (113–115). siG12D-LODER is the only RNAi-based drug
taneous tissue of the scapular region (111). Animals that provides a sustained release of siRNA into the tumor
(males and females) received one or three siG12D- region thanks to a well-known biodegradable PLGA polymer,
LODERs with 0.33 mg siRNA in each implanted de- approved by FDA for drug delivery (116–118). This siRNA
vice, administered on days 1, 14, and 28. Regarding the represents a hopeful strategy against advanced pancreatic can-
safety and toxicity, animals were evaluated for clinical cer. It competes with Atu027 (with a different target) and
signs, weight, ophthalmology, hematology, biochemistry, much lower administered dose (≈ 3 mg) due to the cited
and pathology. For toxicokinetic analysis, blood samples PLGA devices that deliver RNA duplexes with no chemical
were collected after each administration (days 1, 14, and modifications. The formulation into the LODER matrix of
28) at seven temporal points, from 0.25 h to 24 h. PLGA, however, will increase the drug costs. Indeed,
SiG12D-LODER showed safety and tolerability, as no intratumoral administration may has pros and cons.
adverse effect, death, or unscheduled euthanasia oc- Biophase bioavailability is relatively high compared to other
curred. A preclinical finding was a capsule of a thin routes of administration. Also, it is free of infusion-related
fibrotic tissue developed locally in the region of adverse events experienced by patients treated with I.V. for-
LODER implantation. Macrophages and multinucleated mulated siRNAs. However, metastasis of pancreatic cells
giant cells formed a single layer between this capsule will not be controlled by siG12D-LODER unless it was
and the cavity. This reaction did not involve adjacent administered in association with other antineoplastic
tissues and showed no evidence of chronic active inflam- drugs. The procedure for administration the Bpills^ into
mation or necrosis. Finally, only small amounts of pancreatic tumors will require a specialized medical ser-
siG12D siRNA were detected in plasma samples until vice which implies in costs and may reduce the market.
30 min after subcutaneous administration of 3 doses. I.V. route is a comparatively more accessible hospital pro-
A first human phase 1/2a clinical trial evaluated siG12D- cedure. This limitation would be more important whether
LODER in association with chemotherapy for patients with siG12D-LODER implantation must have to be repeated,
non-operable Locally Advanced Pancreatic Cancer (LAPC) but it is not the case. Taking the aggressive behavior of
(NCT01188785) (112). The study examined safety, tolerabil- pancreatic tumor cells, siG12D-LODER is a promising
ity, and antitumor effects. Escalating doses of siG12D- anticancer therapy. Results of a phase 1/2a was positive
LODER in test were 0.025 mg, 0.75 mg (0.375 mg x 2 in a relatively small number of patients, so we must wait
siG12D-LODERs) or 3.0 mg (0.375 mg x 8 siG12D- for an ongoing phase 2 study that is testing siG12DLoder
LODERs) (See Fig. 6b). Some patients received siG12D- (2.8 mg) plus standard chemotherapy for patients with
LODER with a concomitant chemotherapy, like gemcitabine unresectable advanced pancreatic cancer.
or FOLFIRINOX (n = 4). The route of administration is an
outstanding advantage for this RNAi-based drug. By a stan-
dard endoscope ultrasound (EUS) biopsy, SiG12D-LODER’s ANTIMIR IN CLINICAL STUDIES REGISTERED
miniature devices were administered into pancreatic tumor IN NIH
tissue (Fig. 6c). No dose-limiting toxicity occurred, and most
adverse events were grade 1–2. Grade 3–4 adverse reactions Miravirsen
like neutropenia and cholangitis occurred in ten patients
(66.7%). Despite these adverse effects, the drug was consid- Miravirsen (SPC3649) is a microRNA inhibitor designed for
ered safe and well tolerated. SiG12D-LODER showed an treatment of hepatitis C infection (119, 120). Curiously, a
anticancer effect as no patient presented tumor progression. microRNA produced by hepatic cells – microRNA 122
Seven patients also exhibit a decrease in CA19–9 tumor mark- (miR-122), is essential in Hepatitis C Virus (HCV) infection
er, and the median overall survival was ≈ 15 months. A phase (Fig. 7a) (121). A new antiviral drug acting as a miR-122
2 clinical trial (NCT01676259) is now testing the effects of this inhibitor (AntimiR-122) thus has therapeutic potential for this
siRNA in combination with chemotherapy. Patients with liver disease (122). Miravirsen is a 15-base oligonucleotide,
Titze-de-Almeida, David and Titze-de-Almeida

Fig. 7 The role of miR-122 in HCV infection and antiviral action of miravirsen. (a) miR-122 expressed by hepatic cells is an essential host factor for HCV infection,
as it guides AGO2 protein to 5'-UTR region of the virus genome. The binding of Ago2-miR-122 complex to 5'-UTR region protects viral RNA from nucleolytic
degradation, allowing virus propagation in patients with hepatitis C. (b) Miravirsen is an antisense oligo (AntimiR) that binds to and inhibits miR-122 function,
thereby counteracting HCV infection. Chemical changes in miravirsen structure are LNA (red uppercase) and the inclusion of DNA nucleotides (lowercase).
Reprinted with permission from Reference (119).

locked nucleic acid (LNA) modified, phosphorothioated, and placebo over a 29-day period (126). They were followed
complementary to 5′ region of mature miR-122. The newly over 18 weeks for clinical examinations, biochemistry,
formed heteroduplexes miravirsen–miR-122 will prevent the and hematologic tests. Miravirsen caused a significant
binding of miR-122 to viral RNA, compromising HCV rep- dose-dependent and sustained reduction in HCV RNA
lication (Fig. 7b) (119). levels, as measured by a Real-Time HCV assay. The
African green - and cynomolgus monkeys were includ- mean maximum reduction (log10 IU per milliliter) from
ed in preclinical testing on miravirsen activity and safety, baseline reached 1.2, 2.9, and 3.0 for patients receiving
showing positive results (123, 124). Additionally, chronic 3 mg, 5 mg, or 7 mg of miravirsen per kilogram, re-
HCV-infected chimpanzees treated with miravirsen dem- spectively, compared to 0.4 value found in the placebo
onstrated a long-term suppression of viremia and im- group. No dose-limiting adverse events or escape muta-
provement in liver pathology, without viral resistance or tions were found in the miR-122 binding sites of HCV
side effects (125). This AntimiR-122 was allowed to enter sequence. Most adverse events were grade 1. One se-
in phase 1 human clinical trials. vere event occurred, with the loss of consciousness, at
Safety of miravirsen was determined in two phase 1 nine weeks after the last dose of 7 mg/Kg. Two pa-
c l in i c a l t r ia l s ( NC T 00 68 8 01 2, N C T 0 0 97 99 27 ) . tients treated with this dose presented injection-site re-
Miravirsen caused a dose-dependent effect, was well- actions characteristic of oligonucleotide drugs (i.e. ery-
tolerated and showed no dose-limiting toxicities (119). thema, pruritus, burning). Changes in laboratory param-
The drug entered in phase 2 clinical trial that examined eters were not clinically significant to stop miravirsen
safety, tolerability and antiviral effects in subjects with treatment. In summary, this AntimiR-122 oligonucleo-
chronic HCV genotype 1 infection (NCT01200420). tide was well-tolerated and showed antiviral activity in
Patients with plasma HCV RNA level above patients with chronic HCV infection (126).
75,000 IU per milliliter had not received prior antiviral A retrospective analysis of the same patients enrolled in
treatment for HCV genotype 1. Individuals (n = 36) that study (126), evaluated safety and efficacy of miravirsen
received five weekly subcutaneous injections of after an extended period (127). Patients with chronic hepatitis
miravirsen at doses of 3 mg, 5 mg, or 7 mg per kilo- C had received five weekly subcutaneous injections of
gram of body weight (n = 9 patient per dose) or miravirsen or placebo over a period of 29 days. About half
RNAi-based drugs in phase II – III clinical trials

of miravirsen-treated patients underwent treatment with CHALLENGES AND PROGRESS OF RNAI


peginterferon and ribavirin for 3 or 6 weeks and were followed TECHNOLOGY
up for 35 months. This study highlighted the long-term safety
and efficacy of miravirsen. RNAi is the major discovery of the last two decades in life
An in vitro study was aimed at evaluating the ability of sciences. The molecular mechanisms of gene regulation by
miravirsen to act in association with antiviral drugs and po- noncoding RNAs is still a matter of capital importance (7,
tential mutations in viral sequences (128). Miravirsen im- 132). Indeed, RNAi has been very helpful for scientists in a
proved the action of antiviral drugs interferon α2b, ribavirin, more general way, offering a straightforward method to si-
VX-222, 2′-methylcytidine, and telaprevir. However, muta- lence genes of interest. Much information on cell biology
tions in the HCV 5′ untranslated region (5′-UTR) occurred and even the mechanisms of disease are now available thanks
after serial passages (day 72) in vitro. Moreover, mutations were to RNAi techniques (133, 134). As soon as RNAi conquered
also found in samples from subjects that completed a phase 2 its place in research laboratories at the beginning of twenty-
clinical trial (NCT01200420) (128). Association between first century, an exciting question emerged: would this method
miravirsen and telaprevir was further evaluated in phase 1 be transposed from bench to clinics? (135–138). Fortunately, a
clinical trial with healthy volunteers (NCT01646489). decade after the Nobel prize was awarded to Fire and Mello
Finally, miravirsen may represent a therapeutic option for for their discovery of RNAi, ten synthetic RNAi-based drugs
patients with chronic hepatitis C genotype 1 that are null entered in phases 2–3 clinical trials. These represent good
responders to PEGylated interferon alpha plus ribavirin, as candidates to reach the pharmaceutical market in the coming
will be revealed by ongoing clinical trials (NCT02031133, years (79, 106, 134, 139).
NCT01727934, NCT02508090, and NCT01872936). Development of RNAi drugs faces a crucial obstacle: how
to deliver such polar / hydrophilic oligonucleotides across
lipid barriers of the organism? In fact, delivery of nucleic acids
Critical Analysis into cells and tissues is still a matter of concern, but not a
technical barrier for drug development. Three ophthalmic
Miravirsen is the only drug of this review that targets a drugs of this review are naked siRNAs, which means they
microRNA sequence. Most RNAi-based drugs in clinical could transfect cells without the help of any carrier. PF-
testing are siRNAs (not targeting miRNAs) because they 04523655 and QPI-1007 are injected by IVT route, while
represent the first molecules used for gene silencing and SYL040012 is an eye drops. Moreover, naked siRNAs can
experimental therapeutics, soon after the Fire & Melo’s be administered systemically. QPI-1002, a drug used to pre-
striking finding (1, 79). MicroRNAs compose the second vent kidney injury, is given by I.V. route and rapidly accumu-
- but not the less important - Bwave of discovery^ on lates in proximal tubule cells. Kidney and liver are organs that
RNAi, playing a role in many diseases including host- naturally receive the largest amount of oligonucleotides given
pathogen viral interaction (129). Miravirsen is an anti- by this route (30, 31). The anti-HCV miravirsen is another
HCV miR-122 inhibitor. Curiously, miravirsen is ad- example of naked oligonucleotide administered by I.V. route
ministered via subcutaneous injection. This route is suit- properly internalized by liver cells (119). This AntimiR-122 is
able for chronic diseases, as used for insulin injections a short single-stranded antisense DNA nucleotide with two
for example. The rate of absorption is constant and key advantageous changes in chemical structure: LNA which
slow, providing a sustained effect for the weekly injec- increases stability to hybridize with miR-122 and a
tions of miravirsen (126, 130). Regarding structure, phosphorotioated backbone for nuclease resistance (123,
miravirsen is a single stranded antisense DNA oligo, 140). In fact, five weekly injections of miravirsen caused a
containing chemical modifications for stability and spec- long-term and dose-dependent suppression of miR-122 in pa-
ificity. It is administered naked, reducing the costs of tients with hepatitis C (126). Gymnosis is a term coined for
formulation in nanoparticles. That is of capital impor- delivery of short single-stranded oligonucleotides into cells
tance, as patients will receive high quantities of without assistance of transfection agents (141, 142).
miravirsen of ≈ 2400 mg (7 mg/Kg X 70 Kg X 5 TKM-080301, Atu027, and patisiran are not naked
weekly injections) or even higher dosing. Indeed, other siRNAs. Instead, these drugs were complexed with particles
antiviral drugs against HCV are available (131). for delivery of RNAi molecules which are revised elsewhere
Miravirsen showed a dose-dependent reduction in (56, 57, 143, 144). Cationic liposomes (i.e. lipofectamine) are
HCV levels. Antiviral effects of this AntimiR-122 are lipid vesicles commonly used for transfection of siRNAs and
now under evaluation in ongoing phase II studies, to non-viral vectors in different cell types and experimental as-
PEGylated Interferon and antivirus drugs resistant says (145–148). All three drugs are administered by I.V. route,
HCV. Miravirsen is one more option in HCV Roche formulated in lipid particles with technological improvements
treatments. over the conventional liposomes. TKM-080301 uses stable-
Titze-de-Almeida, David and Titze-de-Almeida

nucleic-acid-lipid particles (SNALPs). The major improve- enhanced delivery (CED) is a viable strategy to circum-
ment is a lipid bilayer composed by cationic and fusogenic vent this barrier and administrate RNAi drugs in brain
lipid and coated with polyethylene glycol (PEG). SNALPs areas of interest (154–156).
protect siRNAs from serum nucleases attack while allowing
cellular endosomal uptake for cytoplasmatic release of the
siRNAs (149, 150). PEGylation minimizes phagocytosis and DRUGGING RNAI MOLECULES
avoid renal excretion of siRNAs (134). Arbutus Biopharma
developed its own proprietary delivery system – lipid nano- Inaugurating a new pharmacological drug class is far from an
particle (LNP) platform, which also includes amino lipids and easy matter, besides consuming efforts and a high amount of
cholesterol (151). TKM-080301 reduced tumors located in time and money. The road from bench to clinics has been a
two different organs, liver and adrenal cortex (59, 62). difficult way but not an insurmountable barrier (138). In this
Regarding Atu027, it was formulated in a lipid particle con- scenario, RNAi has shown a quite particular fragmented time-
taining three distinct lipids, AtuFect01 (positively charged), line. First occurred a disproportional optimism soon after the
fusogenic DPhyPE helperlipid (neutral) and MPEG-2000- Fire and Melo’s Nobel Prize in 2006 - called as Birrational
DSPE (PEGylated). This siRNA-lipoplex technology allowed exuberance^ by Haussecker and Kay (157). Then RNAi bio-
a marked uptake of 19-mer siRNA duplexes by the vascular tech suffered a backlash as that huge expectation was not
endothelium in the liver, heart and lung (65, 68, 69). Atu027 achieved (2008 to 2011). Fortunately, the companies entered
showed efficacy against tumors developing in prostate and into a more stable and mature phase of drug development.
pancreas and prevented lymph node lung metastasis in ro- Drugging RNAi is an evolving matter. Advances in the
dents (65, 66). This siRNA was also effective in patients with chemical structure of siRNAs and delivery systems have made
advanced refractory cancer of different organs (i.e. breast, the drugs safe and efficient, as revised elsewhere (139, 158,
pancreas, colon and others) (67). 159). Conjugating siRNAs with specific molecules is a prom-
Tissue-targeted delivery of RNAi therapeutics was a re- ising strategy for a targeted-delivery (160). Among them is N-
markable challenge surpassed by patisiran and siG12D- acetylgalactosamine (GalNAc), a ligand for a receptor found
LODER. Patisiran was designed to knockdown a mutated in hepatocytes. siRNA-GalNac conjugates injected subcuta-
and nonmutated TTR gene that causes amyloidosis (99). neously can trigger a marked knock-down of target mRNA
Beyond a treatment for a genetic disease, this medication in liver. Such technology developed by Alnylam is been used
brought a clear example of a tissue-targeted RNAi delivery in various products (161). This delivery strategy was success-
after I.V. injection. The lipid nanoparticle that carries the fully employed in a product named Revusiran for the treat-
siRNA is opsonized by apolipoprotein E, allowing its binding ment of ATTR amyloidosis (162), the same disease and target
to receptors on hepatocytes. After endocytosis and fusion with gene also addressed by Patisiran that is formulated in lipid
endosomal membranes, the siRNAs are finally released in cell carrier. Revusiran had reached the phase 3 but was
cytoplasm to trigger RNAi on TTR mRNA. Previous at- discontinued by Alnylan in May 2016 due to safety concerns
tempts were also made to deliver nucleic acids into spe- (163). While reinforcing the importance of Monitoring
cific organs. Immunoliposomes coated with monoclonal Committees during ongoing clinical trials, it highlights the
antibodies for binding to receptors of insulin and transfer- vulnerability of any new technological platform.
rin present in blood-brain barrier and neuronal cell mem- Many other promising strategies for delivery and siRNA
brane, were able to deliver nonviral vectors for RNAi into conjugation have been developed, including monoclonal an-
cerebral neurons (152). tibodies and aptamers (164). Monoclonal antibodies are at-
SiG12D-LODER showed how regional administra- tractive molecules for tissue-specific delivery of siRNAs
tion could be used as an innovative way for site- (165). The rationale of this approach looks perfect, but the
specific siRNA delivery. Miniature cylindrical Bpills^ efficacy is still limited (166). Both the cellular uptake and the
made with the biocompatible polymer PLGA were im- following cytoplasm trafficking and siRNA delivery are issues
planted regionally into pancreatic tumors by endoscope that may compromise the success of such strategy (134). A
ultrasound (EUS) biopsy for a long term delivery of recent work presented the named THIOMAB platform for
KRAS-targeted siRNAs, showing anticancer effects antibody-siRNA conjugates delivery. Although offering po-
(109, 112). Regional application of RNAi-based thera- tential improvements in conjugation, the method revealed
peutics is a viable route to treat diseases in organs of some limitations about cell receptors capable of internalizing
difficult access. Devices like catheters or stents for local siRNAs, intracellular trafficking and endosomal escape (167).
injections might circumvent the delivery of nucleic acids Regarding aptamers, these are synthetic oligonucleotides se-
in such tissues (120). Some systemic therapeutics for lected to bind with high affinity to receptors (168). RNA
brain diseases may also fail because of the blood-brain aptamers can be conjugated with siRNAs for a targeted
barrier (153). In this sense, the method of convection- delivery (169, 170). A critical point is the selection of
RNAi-based drugs in phase II – III clinical trials

receptors that efficiently internalize cargo to cell cyto- 8. Ketting RF. The many faces of RNAi. Dev Cell. 2011;20(2):148–
61.
plasm, as addressed by a new methodology named Bcell-
9. Pratt AJ, MacRae IJ. The RNA-induced silencing complex: a
internalization SELEX^ (171). versatile gene-silencing machine. J Biol Chem. 2009;284(27):
Research on mechanisms of siRNA internalization in tar- 17897–901.
get cells and further trafficking, as well as the development of 10. Jinek M, Doudna JA. A three-dimensional view of the molecular
machinery of RNA interference. Nature. 2009;457(7228):405–12.
ligands and delivery systems are matters of capital importance
11. Zamore PD, Tuschl T, Sharp PA, Bartel DP. RNAi: double-
in RNAi biotechnology. Any system for oligonucleotide deliv- stranded RNA directs the ATP-dependent cleavage of mRNA at
ery, either using a nanoparticle or a conjugative molecule, will 21 to 23 nucleotide intervals. Cell. 2000;101(1):25–33.
present pros and cons regarding cost, specificity, efficiency, 12. Meister G, Tuschl T. Mechanisms of gene silencing by double-
stranded RNA. Nature. 2004;431(7006):343–9.
and even toxicity.
13. Brafman A, Mett I, Shafir M, Gottlieb H, Damari G, Gozlan-
Kelner S, Vishnevskia-Dai V, Skaliter R, Einat P, Faerman A,
Feinstein E, Shoshani T. Inhibition of oxygen-induced retinopa-
thy in RTP801-deficient mice. Invest Ophthalmol Vis Sci.
SUMMARY 2004;45(10):3796–805.
14. Rittenhouse KD, Hirakawa, B., Huang, W., Basile, A.S., Johnson,
Synthetic RNAi-based drugs have shown ability to silence T.R., Shachar R.A. Dose-related gene silencing of RTP801 with
different genes, acting in diseases with distinct pathogenesis, the siRNA PF04523655 in Long Evans rat models of STZ in-
duced diabetes and laser induced CNV [abstract]. Invest
from cell death by apoptosis to those caused by uncontrolled Ophthalmol Vis Sci. 2010;51:E-abstract 6447.
cell proliferation. The established diseases for RNAi therapeu- 15. Rittenhouse KD, Kalabat, D., Yang, A., Vicini, P., Johnson, T.R.,
tics are: 1- death of kidney cells after injury by ischemia, cis- Huang, W., Hirakawa, B.; Basile, A. S.; Schachar, R.A.
platin or transplantation (QPI-1002); 2- tumor growth in dif- Characterization of regional RTP801 gene expression within the
retina and the concentration-effect relationship of PF-655, an
ferent organs, and lung metastasis (TKM-080301, Atu027, RTP801-silencing siRNA, following intravitreous administration
siG12D-LODER); 3- HCV genotype 1 infection in chronic to diabetic rats [abstract]. Invest Ophthalmol Vis Sci. 2011;52:E-
hepatitis C (miravirsen); 4- ophthalmic disorders, including Abstract 5641.
AMD, NAION, glaucoma, and ocular pain (PF-04523655, 16. Nguyen QD, Schachar RA, Nduaka CI, Sperling M, Klamerus
KJ, Chi-Burris K, Yan E, Paggiarino DA, Rosenblatt I, Aitchison
QPI-1007, SYL040012, SYL1001); 5- transthyretin-mediated R, Erlich SS, Group MCS. Evaluation of the siRNA PF-
amyloidosis (patisiran). Clinical trials revealed these products 04523655 versus ranibizumab for the treatment of neovascular
are safe and well tolerated, as most adverse effects were mild age-related macular degeneration (MONET study).
to moderate, and occurred during drug injection, either local- Ophthalmology. 2012;119(9):1867–73.
17. Rittenhouse KD, Johnson TR, Vicini P, Hirakawa B, Kalabat D,
ly or systemically. Infusion-related reactions after I.V. injec- Yang AH, Huang W, Basile AS. RTP801 gene expression is dif-
tions are transient and could be prevented by glucocorticoids, ferentially upregulated in retinopathy and is silenced by PF-
antihistamines and others. In conclusion, RNAi therapeutic 04523655, a 19-mer siRNA directed against RTP801. Invest
drugs will take place in the pharmaceutical market in the Ophthalmol Vis Sci. 2014;55(3):1232–40.
18. Nguyen QD, Schachar RA, Nduaka CI, Sperling M, Basile AS,
few coming years. Klamerus KJ, Chi-Burris K, Yan E, Paggiarino DA, Rosenblatt I,
Khan A, Aitchison R, Erlich SS, Group PFS. Phase 1 dose-
escalation study of a siRNA targeting the RTP801 gene in age-
related macular degeneration patients. Eye (London, England).
2012;26(8):1099–105.
REFERENCES 19. Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK,
Chung CY, Kim RY, Group MS. Ranibizumab for
neovascular age-related macular degeneration. N Engl J
1. Bobbin ML, Rossi JJ. RNA interference (RNAi)-based therapeu-
Med. 2006;355(14):1419–31.
tics: delivering on the promise? Annu Rev Pharmacol Toxicol.
2016;56:103–22. 20. Feinstein E, Ashush, H., Kleinman, M.E. et al. PF-0452655
[REDD14], an siRNA compound targeting RTP801, penetrates
2. Martinez T, Gonzalez MV, Roehl I, Wright N, Paneda C,
retinal cells producing target gene knockdown and avoiding
Jimenez AI. In vitro and in vivo efficacy of SYL040012, a novel
TLR3 activation. Poster Present Assoc Res Vis Ophthalmol
siRNA compound for treatment of glaucoma. Mol Ther.
Annu Meet May 7, Ft Lauderdale, FL. 2009.
2014;22(1):81–91.
21. Levenson JH, Kozarsky, A.. Visual acuity. In: Walker HK, Hall
3. Hannon GJ. RNA interference. Nature. 2002;418(6894):244–51. W.D., Hurst J.W., editor. Clinical methods, the history, physical,
4. Hammond SM, Caudy AA, Hannon GJ. Post- and laboratory examinations. Boston: Butterworths; 1990.
transcriptional gene silencing by double-stranded RNA. 22. Enslow R, Bhuvanagiri S, Vegunta S, Cutler B, Neff M, Stagg B.
Nat Rev Genet. 2001;2(2):110–9. Association of Anti-VEGF injections with progression of geo-
5. Carthew RW, Sontheimer EJ. Origins and mechanisms of graphic atrophy. Ophthalmol Eye Dis. 2016;8:31–2.
miRNAs and siRNAs. Cell. 2009;136(4):642–55. 23. Gemenetzi M, Lotery AJ, Patel PJ. Risk of geographic atrophy in
6. Bushati N, Cohen SM. microRNA functions. Annu Rev Cell Dev age-related macular degeneration patients treated with intravitre-
Biol. 2007;23:175–205. al anti-VEGF agents. Eye (London, England) 2017;31(1):1–9.
7. Svoboda P. Renaissance of mammalian endogenous RNAi. FEBS 24. Amadio M, Govoni S, Pascale A. Targeting VEGF in eye neovas-
Lett. 2014;588(15):2550–6. cularization: What's new?: a comprehensive review on current
Titze-de-Almeida, David and Titze-de-Almeida

therapies and oligonucleotide-based interventions under develop- 42. Kisiswa L, Dervan AG, Albon J, Morgan JE, Wride MA. Retinal
ment. Pharmacol Res. 2016;103:253–69. ganglion cell death postponed: giving apoptosis a break?
25. Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet. Ophthalmic Res. 2010;43(2):61–78.
2012;380(9843):756–66. 43. Levin LA. Axonal loss and neuroprotection in optic neuropathies.
26. Brown JR, Rezaee ME, Marshall EJ, Matheny ME. Hospital Can J Ophthalmol. 2007;42(3):403–8.
mortality in the United States following acute kidney injury. 44. Schmidt KG, Bergert H, Funk RH. Neurodegenerative diseases of
Biomed Res Int. 2016;2016:4278579. the retina and potential for protection and recovery. Curr
27. Hoste EA, Schurgers M. Epidemiology of acute kidney injury: Neuropharmacol. 2008;6(2):164–78.
how big is the problem? Crit Care Med. 2008;36(4 Suppl): 45. Kurokawa T, Katai N, Shibuki H, Kuroiwa S, Kurimoto Y,
S146–51. Nakayama C, Yoshimura N. BDNF diminishes caspase-2 but
28. Bonegio R, Lieberthal W. Role of apoptosis in the pathogenesis of not c-Jun immunoreactivity of neurons in retinal ganglion cell
acute renal failure. Curr Opin Nephrol Hypertens. 2002;11(3): layer after transient ischemia. Invest Ophthalmol Vis Sci.
301–8. 1999;40(12):3006–11.
29. Molitoris BA, Dagher PC, Sandoval RM, Campos SB, Ashush H, 46. Singh M, Savitz SI, Hoque R, Gupta G, Roth S, Rosenbaum PS,
Fridman E, Brafman A, Faerman A, Atkinson SJ, Thompson JD, Rosenbaum DM. Cell-specific caspase expression by different
Kalinski H, Skaliter R, Erlich S, Feinstein E. siRNA targeted to neuronal phenotypes in transient retinal ischemia. J Neurochem.
p53 attenuates ischemic and cisplatin-induced acute kidney injury. 2001;77(2):466–75.
J Am Soc Nephrol. 2009;20(8):1754–64. 47. Vigneswara V, Berry M, Logan A, Ahmed Z. Pharmacological
30. Braasch DA, Paroo Z, Constantinescu A, Ren G, Oz OK, Mason inhibition of caspase-2 protects axotomised retinal ganglion cells
RP, Corey DR. Biodistribution of phosphodiester and phospho- from apoptosis in adult rats. PLoS One. 2012;7(12):e53473.
rothioate siRNA. Bioorg Med Chem Lett. 2004;14(5):1139–43. 48. Ahmed Z, Kalinski H, Berry M, Almasieh M, Ashush H, Slager N,
31. Agrawal S, Temsamani J, Galbraith W, Tang J. Brafman A, Spivak I, Prasad N, Mett I, Shalom E, Alpert E, Di
Pharmacokinetics of antisense oligonucleotides. Clin Polo A, Feinstein E, Logan A. Ocular neuroprotection by siRNA
Pharmacokinet. 1995;28(1):7–16. targeting caspase-2. Cell Death Dis. 2011;2:e173.
32. Garin D, Ahmadi M, Gauchez AS, Bohic S, Boccard S, Cloetens 49. Solano EC, Kornbrust DJ, Beaudry A, Foy JW, Schneider DJ,
P, Fagret D, Berger F, Briat A, Ghezzi C, Pelletier L. In vivo Thompson JD. Toxicological and pharmacokinetic properties of
siRNA distribution and pharmacokinetics assessed by nuclear im- QPI-1007, a chemically modified synthetic siRNA targeting cas-
aging are modulated according to radiolabelling site. Nucl Med pase 2 mRNA, following intravitreal injection. Nucleic Acid
Biol. 2015;42(12):958–66. Therapeutics. 2014;24(4):258–66.
33. Sands H, Gorey-Feret LJ, Cocuzza AJ, Hobbs FW, Chidester D, 50. Vigneswara V, Ahmed Z. Long-term neuroprotection of retinal
Trainor GL. Biodistribution and metabolism of internally 3H- ganglion cells by inhibiting caspase-2. Cell Death Dis. 2016;2:
labeled oligonucleotides. I. Comparison of a phosphodiester and 16044.
a phosphorothioate. Mol Pharmacol. 1994;45(5):932–43. 51. [Internet]. QP. Quark Pharmaceuticals Doses First Non-Arteritic
34. van de Water FM, Boerman OC, Wouterse AC, Peters JG, Russel Anterior Ischemic Optic Neuropathy (NAION) Patient in
FG, Masereeuw R. Intravenously administered short interfering Stratum II of its Clinical Study of siRNA Drug Candidate QPI-
RNA accumulates in the kidney and selectively suppresses gene 1007. 2010 News release October 20, 2010. Available from:
function in renal proximal tubules. Drug Metab Dispos. http://quarkpharma.com/?p=11968.
2006;34(8):1393–7. 52. [Internet]. QP. Quark Announces First Patient Dosed in a Phase
35. Viel T, Boisgard R, Kuhnast B, Jego B, Siquier-Pernet K, Hinnen IIa Trial Evaluating QPI-1007 for Neuroprotection in Patients
F, Dolle F, Tavitian B. Molecular imaging study on in vivo distri- with Acute Primary Angle Closure Glaucoma. 2014 News release,
bution and pharmacokinetics of modified small interfering RNAs June 25, 2014. Available from: http://quarkpharma.
(siRNAs). Oligonucleotides. 2008;18(3):201–12. com/?p=11625.
36. Imamura R, Isaka Y, Sandoval RM, Ori A, Adamsky S, Feinstein 53. [Internet] QP. Quark Pharmaceuticals Awarded Key Patent for
E, Molitoris BA, Takahara S. Intravital two-photon microscopy QPI-1007 Ocular Neuroprotectant. 2016 News release July 28,
assessment of renal protection efficacy of siRNA for p53 in exper- 2016. Avaeilable from: http://quarkpharma.com/?p=12340.
imental rat kidney transplantation models. Cell Transplant. 54. Malumbres M, Barbacid M. Cell cycle, CDKs and cancer: a
2010;19(12):1659–70. changing paradigm. Nat Rev Cancer. 2009;9(3):153–66.
37. Thompson JD, Kornbrust DJ, Foy JW, Solano EC, Schneider DJ, 55. Liu X. Targeting polo-like kinases: a promising therapeutic ap-
Feinstein E, Molitoris BA, Erlich S. Toxicological and pharmaco- proach for cancer treatment. Transl Oncol. 2015;8(3):185–95.
kinetic properties of chemically modified siRNAs targeting p53 56. Alabi C, Vegas A, Anderson D. Attacking the genome: emerging
RNA following intravenous administration. Nucleic Acid siRNA nanocarriers from concept to clinic. Curr Opin
Therapeutics. 2012;22(4):255–64. Pharmacol. 2012;12(4):427–33.
38. Siedlecki A, Irish W, Brennan DC. Delayed graft function in the 57. Kanasty R, Dorkin JR, Vegas A, Anderson D. Delivery materials
kidney transplant. Am J Transplant. 2011;11(11):2279–96. for siRNA therapeutics. Nat Mater. 2013;12(11):967–77.
39. Perico N, Cattaneo D, Sayegh MH, Remuzzi G. Delayed 58. Semple SC, Judge, A.D., Robbins, M., Klimuk, S., Eisenhardt,
g r a f t f u nc ti o n in k i d n e y tr a n s p l a n t a ti o n . L a n c e t . M., Crosley, E., et al. Preclinical characterization of TKM-
2004;364(9447):1814–27. 080301, a lipid nanoparticle formulation of a small interfering
40. [Internet] QP. Quark Pharmaceuticals Reports Favorable Results RNA directed against polo-like kinase 1 [abstract]. Cancer Res
from Phase II Clinical Trial Evaluating Investigational siRNA 2011;71 Suppl 8:Abstract nr 2829.
QPI-1002. 2014 News release July 28, 2014. Available from: 59. Ramanathan RK, Hamburg, S.I., Borad, M.J., Seetharam, M.,
http://quarkpharma.com/?p=11621. Kundranda, M.N., Lee, P., et al. A phase I dose escalation study of
41. [Internet]. QP. Quark Pharmaceuticals and Major TKM-080301, a RNAi therapeutic directed against PLK1, in
Pharmaceutical Company Enter into Licensing Option patients with advanced solid tumors [abstract]. Cancer Res
Agreement for the p53 Suppressor Drug QPI-1002, the First 2013;73 (Suppl 8):Abstract nr LB-289.
siRNA Administered Systemically in Human. 2010 News release 60. [Internet]. TPC. Tekmira Presents Data From Its TKM-PLK1
August 18, 2010. Phase I Clinical Trial at American Association for Cancer
RNAi-based drugs in phase II – III clinical trials

Research (AACR) Meeting April 9, 2013. Available from: 75. Weinreb RN, Khaw PT. Primary open-angle glaucoma. Lancet.
https://globenewswire.com/news-release/2013/04/09/537016 2004;363(9422):1711–20.
/10027901/en/Tekmira-Presents-Data-From-Its-TKM-PLK1- 76. Weinreb RN, Aung T, Medeiros FA. The pathophysiology and
Phase-I-Clinical-Trial-at-American-Association-for-Cancer- treatment of glaucoma: a review. J Am Med Assoc. 2014;311(18):
Research-AACR-Meeting.html. 1901–11.
61. Northfelt D.W. H, S.I., Borad, M.J., Seetharam, M., Curtis, K.K., 77. Dejneka NS, Wan S, Bond OS, Kornbrust DJ, Reich SJ. Ocular
Lee, P., et al. A phase I dose-escalation study of TKM-080301, a biodistribution of bevasiranib following a single intravitreal injec-
RNAi therapeutic directed against polo-like kinase 1 (PLK1), in tion to rabbit eyes. Mol Vis. 2008;14:997–1005.
patients with advanced solid tumors: Expansion cohort evaluation 78. Kleinman ME, Yamada K, Takeda A, Chandrasekaran V,
of biopsy samples for evidence of pharmacodynamic effects of Nozaki M, Baffi JZ, Albuquerque RJ, Yamasaki S, Itaya M, Pan
PLK1 inhibition [abstract]. J Clin Oncol 2013;31 Suppl:abstr Y, Appukuttan B, Gibbs D, Yang Z, Kariko K, Ambati BK,
TPS2621. Wilgus TA, DiPietro LA, Sakurai E, Zhang K, Smith JR,
62. Demeure MJ, Armaghany, T., Ejadi, S., Ramanathan, R.K., Taylor EW, Ambati J. Sequence- and target-independent angio-
Elfiky, A., Jonathan R. Strosberg, J.R., et al.. A phase I/II study genesis suppression by siRNA via TLR3. Nature. 2008;452(7187):
of TKM-080301, a PLK1-targeted RNAi in patients with adre- 591–7.
nocortical cancer (ACC) [abstract]. J Clin Oncol 2016;34 Suppl: 79. Burnett JC, Rossi JJ. RNA-based therapeutics: current progress
abstr 2547. and future prospects. Chem Biol. 2012;19(1):60–71.
63. Gutteridge RE, Ndiaye MA, Liu X, Ahmad N. Plk1 inhibitors in 80. Campochiaro PA. Potential applications for RNAi to probe path-
cancer therapy: from laboratory to clinics. Mol Cancer Ther. ogenesis and develop new treatments for ocular disorders. Gene
2016;15(7):1427–35. Ther. 2006;13(6):559–62.
64. Unsal-Kacmaz K, Ragunathan S, Rosfjord E, Dann S, Upeslacis 81. Elena PP, Kosina-Boix M, Moulin G, Lapalus P.
E, Grillo M, Hernandez R, Mack F, Klippel A. The interaction of Autoradiographic localization of beta-adrenergic receptors in
PKN3 with RhoC promotes malignant growth. Mol Oncol. rabbit eye. Invest Ophthalmol Vis Sci. 1987;28(8):1436–41.
2012;6(3):284–98. 82. Moreno-Montanes J, Sadaba B, Ruz V, Gomez-Guiu A, Zarranz
65. Aleku M, Schulz P, Keil O, Santel A, Schaeper U, Dieckhoff B, J, Gonzalez MV, Paneda C, Jimenez AI. Phase I clinical trial of
Janke O, Endruschat J, Durieux B, Roder N, Loffler K, Lange C, SYL040012, a small interfering RNA targeting beta-adrenergic
Fechtner M, Mopert K, Fisch G, Dames S, Arnold W, Jochims K, receptor 2, for lowering intraocular pressure. Mol Ther.
Giese K, Wiedenmann B, Scholz A, Kaufmann J. Atu027, a lipo- 2014;22(1):226–32.
somal small interfering RNA formulation targeting protein kinase 83. Gonzalez V, Palumaa K, Turman K, Muñoz FJ, Jordan J, García
N3, inhibits cancer progression. Cancer Res. 2008;68(23):9788– J, Ussa F, Antón A, Gutierrez E, Moreno-Montanes J. Phase 2 of
98. bamosiran (SYL040012), a novel RNAi based compound for the
treatment of increased intraocular pressure associated to glauco-
66. Santel A, Aleku M, Roder N, Mopert K, Durieux B, Janke O,
Keil O, Endruschat J, Dames S, Lange C, Eisermann M, Loffler ma [abstract]. Invest Ophthalmol Vis Sci. 2014;55:564.
K, Fechtner M, Fisch G, Vank C, Schaeper U, Giese K, 84. Gonzalez V, Moreno-Montanes J, Oll M, Sall KN, Palumaa K,
Kaufmann J. Atu027 prevents pulmonary metastasis in experi- Dubiner H, Turman K, Muñoz-Negrete F, Ruz V, Jimenez AI.
mental and spontaneous mouse metastasis models. Clin Cancer Results of Phase IIB SYLTAG clinical trial with bamosiran in
Res. 2010;16(22):5469–80. patients with glaucoma [abstract]. Investig Ophthalmol Vis Sci.
2016;57:3023.
67. Schultheis B, Strumberg D, Santel A, Vank C, Gebhardt F, Keil
85. [Internet] S. Sylentis announces top-line results from Phase II
O, Lange C, Giese K, Kaufmann J, Khan M, Drevs J. First-in-
study SYLTAG for RNAi drug bamosiran (SYL040012) in
human phase I study of the liposomal RNA interference therapeu-
Glaucoma. 2015.
tic Atu027 in patients with advanced solid tumors. J Clin Oncol.
86. Muller ME, van der Velde N, Krulder JW, van der Cammen TJ.
2014;32(36):4141–8.
Syncope and falls due to timolol eye drops. BMJ. 2006;332(7547):
68. Strumberg D, Schultheis B, Traugott U, Vank C, Santel A, Keil
960–1.
O, Giese K, Kaufmann J, Drevs J. Phase I clinical development of
87. Maenpaa J, Pelkonen O. Cardiac safety of ophthalmic timolol.
Atu027, a siRNA formulation targeting PKN3 in patients with
Expert Opin Drug Saf. 2016;15(11):1549–61.
advanced solid tumors. Int J Clin Pharmacol Ther. 2012;50(1):
88. Vander Zanden JA, Valuck RJ, Bunch CL, Perlman JI, Anderson
76–8.
C, Wortman GI. Systemic adverse effects of ophthalmic beta-
69. Santel A, Aleku M, Keil O, Endruschat J, Esche V, Fisch G, blockers. Ann Pharmacother. 2001;35(12):1633–7.
Dames S, Loffler K, Fechtner M, Arnold W, Giese K, Klippel 89. Szallasi A, Cortright DN, Blum CA, Eid SR. The vanilloid recep-
A, Kaufmann J. A novel siRNA-lipoplex technology for RNA tor TRPV1: 10 years from channel cloning to antagonist proof-of-
interference in the mouse vascular endothelium. Gene Ther. concept. Nat Rev Drug Discov. 2007;6(5):357–72.
2006;13(16):1222–34. 90. Gonzalez V, Jímenez AI, Martínez T. Targeting TRPV1 receptor
70. [Internet] ST. Atu027 update. 5 April 2016. Available from: for the treatment of ocular pain associated to dry eye syndrome
http://silence-therapeutics-com.s3-eu-west-1.amazonaws. [abstract]. Invest Ophthalmol Vis Sci. 2011;52(14):3844.
com/app/uploads/2016/04/05064530/Atu027.pdf. 91. Ruz V, Gonzalez V, Martinez-Garcia C, Pañeda C, Jiménez AI.
71. Conroy T, Bachet JB, Ayav A, Huguet F, Lambert A, Caramella SYL1001, a new treatment based on RNAi for the treatment of
C, Marechal R, Van Laethem JL, Ducreux M. Current standards ocular pain [abstract]. Invest Ophthalmol Vis Sci. 2014;55(13):
and new innovative approaches for treatment of pancreatic can- 3673.
cer. Eur J Cancer. 2016;57:10–22. 92. Gonzalez V, Moreno-Montañé J, Sádaba B, Ruz V, Jímenez AI.
72. Ansari D, Gustafsson A, Andersson R. Update on the manage- SYL1001 for treatment of ocular discomfort in dry eye: safety and
ment of pancreatic cancer: surgery is not enough. World J tolerance (phase I study) [abstract]. Invest Ophthalmol Vis Sci.
Gastroenterol. 2015;21(11):3157–65. 2012;53(14):575.
73. Quigley HA. Glaucoma Lancet. 2011;377(9774):1367–77. 93. Jiménez AI, Castilho JMB, Moreno-Montanes J, Jimenez-Alfaro I,
74. Coleman AL, Miglior S. Risk factors for glaucoma onset and Muñoz-Negrete F, Palumaa K, Turman K, Paneda C, Martinez
progression. Surv Ophthalmol. 2008;53 Suppl1:S3–10. T, Ruz V, Gonzalez V. Results of clinical trials with a novel RNA-
Titze-de-Almeida, David and Titze-de-Almeida

based therapy (SYL1001) to treat patients with ocular pain asso- 106. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello
ciated to dry eye disease. Invest Ophthalmol Vis Sci. 2016;57(12): CC. Potent and specific genetic interference by double-stranded
2878. RNA in Caenorhabditis elegans. Nature. 1998;391(6669):806–11.
94. Milner MS, Beckman KA, Luchs JI, Allen QB, Awdeh RM, 107. Adams D, Cauquil C, Labeyrie C, Beaudonnet G, Algalarrondo
Berdahl J, Boland TS, Buznego C, Gira JP, Goldberg DF, V, Theaudin M. TTR kinetic stabilizers and TTR gene silencing:
Goldman D, Goyal RK, Jackson MA, Katz J, Kim T, a new era in therapy for familial amyloidotic polyneuropathies.
Majmudar PA, Malhotra RP, MB MD, Rajpal RK, Raviv T, Expert Opin Pharmacother. 2016;17(6):791–802.
Rowen S, Shamie N, Solomon JD, Stonecipher K, Tauber S, 108. Kerschen P, Plante-Bordeneuve V. Current and future treatment
Trattler W, Walter KA, GOt W, Weinstock RJ, Wiley WF, Yeu approaches in transthyretin familial amyloid polyneuropathy.
E. Dysfunctional tear syndrome: dry eye disease and associated Curr Treat Options Neurol. 2016;18(12):53.
tear film disorders - new strategies for diagnosis and treatment. 109. Zorde Khvalevsky E, Gabai R, Rachmut IH, Horwitz E,
Curr Opin Ophthalmol. 2017;27(Suppl 1):3–47. Brunschwig Z, Orbach A, Shemi A, Golan T, Domb AJ, Yavin
95. Al-Saedi Z, Zimmerman A, Bachu RD, Dey S, Shah Z, Baugh R, E, Giladi H, Rivkin L, Simerzin A, Eliakim R, Khalaileh A,
Boddu SH. Dry eye disease: present challenges in the manage- Hubert A, Lahav M, Kopelman Y, Goldin E, Dancour A,
ment and future trends. Curr Pharm Des. 2016;22(28):4470–90. Hants Y, Arbel-Alon S, Abramovitch R, Shemi A, Galun E.
96. Sekijima Y. Recent progress in the understanding and Mutant KRAS is a druggable target for pancreatic cancer. Proc
treatment of transthyretin amyloidosis. J Clin Pharm Natl Acad Sci U S A. 2013;110(51):20723–8.
Ther. 2014;39(3):225–33. 110. Shemi A, Khvalevsky EZ, Gabai RM, Domb A, Barenholz Y.
97. Hawkins PN, Ando Y, Dispenzeri A, Gonzalez-Duarte A, Adams Multistep, effective drug distribution within solid tumors.
D, Suhr OB. Evolving landscape in the management of Oncotarget. 2015;6(37):39564–77.
transthyretin amyloidosis. Ann Med. 2015;47(8):625–38. 111. Ramot Y, Rotkopf S, Gabai RM, Zorde Khvalevsky E, Muravnik
98. Adams D, Suhr OB, Hund E, Obici L, Tournev I, Campistol JM, S, Marzoli GA, Domb AJ, Shemi A, Nyska A. Preclinical safety
Slama MS, Hazenberg BP, Coelho T. European Network for T- evaluation in rats of a polymeric matrix containing an siRNA drug
F. First European consensus for diagnosis, management, and used as a local and prolonged delivery system for pancreatic can-
treatment of transthyretin familial amyloid polyneuropathy. cer therapy. Toxicol Pathol. 2016;44(6):856–65.
Curr Opin Neurol. 2016;29(Suppl 1):S14–26. 112. Golan T, Khvalevsky EZ, Hubert A, Gabai RM, Hen N, Segal A,
99. Coelho T, Adams D, Silva A, Lozeron P, Hawkins PN, Mant T, Domb A, Harari G, David EB, Raskin S, Goldes Y, Goldin E,
Perez J, Chiesa J, Warrington S, Tranter E, Munisamy M, Eliakim R, Lahav M, Kopleman Y, Dancour A, Shemi A, Galun
Falzone R, Harrop J, Cehelsky J, Bettencourt BR, Geissler M, E. RNAi therapy targeting KRAS in combination with chemo-
Butler JS, Sehgal A, Meyers RE, Chen Q, Borland T, therapy for locally advanced pancreatic cancer patients.
Hutabarat RM, Clausen VA, Alvarez R, Fitzgerald K, Gamba- Oncotarget. 2015;6(27):24560–70.
Vitalo C, Nochur SV, Vaishnaw AK, Sah DW, Gollob JA, Suhr 113. Hunter JC, Gurbani D, Ficarro SB, Carrasco MA, Lim SM, Choi
OB. Safety and efficacy of RNAi therapy for transthyretin amy- HG, Xie T, Marto JA, Chen Z, Gray NS, Westover KD. In situ
loidosis. N Engl J Med. 2013;369(9):819–29. selectivity profiling and crystal structure of SML-8-73-1, an active
100. Szebeni J, Muggia F, Gabizon A, Barenholz Y. Activation of com- site inhibitor of oncogenic K-Ras G12C. Proc Natl Acad Sci U S
plement by therapeutic liposomes and other lipid excipient-based A. 2014;111(24):8895–900.
therapeutic products: prediction and prevention. Adv Drug Deliv 114. Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific
Rev. 2011;63(12):1020–30. inhibitors inactivate mutant KRAS G12C by a trapping mecha-
101. Suhr OB, Coelho T, Buades J, Pouget J, Conceicao I, Berk J, nism. Science. 2016;351(6273):604–8.
Schmidt H, Waddington-Cruz M, Campistol JM, Bettencourt 115. Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-
BR, Vaishnaw A, Gollob J, Adams D. Efficacy and safety of Ras(G12C) inhibitors allosterically control GTP affinity and effec-
patisiran for familial amyloidotic polyneuropathy: a phase II tor interactions. Nature. 2013;503(7477):548–51.
multi-dose study. Orphanet Journal of Rare Diseases. 2015;10: 116. Singha K, Namgung R, Kim WJ. Polymers in small-interfering
109. RNA delivery. Nucleic Acid Therapeutics. 2011;21(3):133–47.
102. Koike H, Tanaka F, Hashimoto R, Tomita M, Kawagashira Y, 117. Lu JM, Wang X, Marin-Muller C, Wang H, Lin PH, Yao Q,
Iijima M, Fujitake J, Kawanami T, Kato T, Yamamoto M, Sobue Chen C. Current advances in research and clinical applications
G. Natural history of transthyretin Val30Met familial amyloid of PLGA-based nanotechnology. Expert Rev Mol Diagn.
polyneuropathy: analysis of late-onset cases from non-endemic 2009;9(4):325–41.
areas. J Neurol Neurosurg Psychiatry. 2012;83(2):152–8. 118. Kapoor DN, Bhatia A, Kaur R, Sharma R, Kaur G, Dhawan S.
103. Coelho T, Suhr OB, Conceicao I, Waddington-Cruz M, Schmidt PLGA: a unique polymer for drug delivery. Ther Deliv. 2015;6(1):
H, Buades J, Campistol JM, Pouget J, Berk J, Falzone R, White L, 41–58.
Bettencourt B, Cehelsky J, Nochur S, Vaishnaw A, Gollob J, 119. Lindow M, Kauppinen S. Discovering the first microRNA-
Adams D. Phase 2 open-label extension study of patisiran, an targeted drug. J Cell Biol. 2012;199(3):407–12.
investigational RNAi therapeutic for the treatment of familial am- 120. van Rooij E, Kauppinen S. Development of microRNA
yloid polyneuropathy [abstract]. Abstract [S9003] presented at therap eu tics is coming of ag e. EMBO Molecular
67th Annual Meeting of the American Academy of Neurology. Medicine. 2014;6(7):851–64.
2015. 121. Bandiera S, Pfeffer S, Baumert TF, Zeisel MB. miR-122–a key
104. [Internet] AP. Updates from Patisiran and Revusiran, in factor and therapeutic target in liver disease. J Hepatol.
Development for the Treatment of hATTR Amyloidosis 2015;62(2):448–57.
Available from: http://www.alnylam. 122. Thibault PA, Wilson JA. Targeting miRNAs to treat hepatitis C
com/capella/presentations/updates-from-patisiran-and- virus infections and liver pathology: inhibiting the virus and alter-
revusiran-for-hattr-amyloidosis/. ing the host. Pharmacol Res. 2013;75:48–59.
105. Friedmann T, Roblin R. Gene therapy for human genetic disease? 123. Elmen J, Lindow M, Schutz S, Lawrence M, Petri A, Obad S,
Science. 1972;175(4025):949–55. Lindholm M, Hedtjarn M, Hansen HF, Berger U, Gullans S,
Kearney P, Sarnow P, Straarup EM, Kauppinen S. LNA-
RNAi-based drugs in phase II – III clinical trials

mediated microRNA silencing in non-human primates. Nature. oligonucleotides, unassisted by transfection reagents. Nucleic
2008;452(7189):896–9. Acids Res. 2010;38(1):e3.
124. Hildebrandt-Eriksen ES, Aarup V, Persson R, Hansen HF, Munk 142. Soifer HS, Koch T, Lai J, Hansen B, Hoeg A, Oerum H, Stein
ME, Orum H. A locked nucleic acid oligonucleotide targeting CA. Silencing of gene expression by gymnotic delivery of antisense
microRNA 122 is well-tolerated in cynomolgus monkeys. oligonucleotides. Methods Mol Biol. 2012;815:333–46.
Nucleic Acid Therapeutics. 2012;22(3):152–61. 143. Zhao J, Feng SS. Nanocarriers for delivery of siRNA and co-
125. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow delivery of siRNA and other therapeutic agents. Nanomedicine
M, Munk ME, Kauppinen S, Orum H. Therapeutic silencing of (London, England). 2015;10(14):2199–228.
microRNA-122 in primates with chronic hepatitis C virus infec- 144. Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR,
tion. Science. 2010;327(5962):198–201. Anderson DG. Non-viral vectors for gene-based therapy. Nat
126. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez- Rev Genet. 2014;15(8):541–55.
Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou 145. Sales TT, Resende FF, Chaves NL, Titze-De-Almeida SS, Bao
Y, Persson R, King BD, Kauppinen S, Levin AA, Hodges MR. SN, Brettas ML, Titze-De-Almeida R. Suppression of the Eag1
Treatment of HCV infection by targeting microRNA. N Engl J potassium channel sensitizes glioblastoma cells to injury caused by
Med. 2013;368(18):1685–94. temozolomide. Oncol Lett. 2016;12(4):2581–9.
127. van der Ree MH, van der Meer AJ, de Bruijne J, Maan R, van 146. Castania V, Issy AC, Silveira JW, Ferreira FR, Titze-de-
Vliet A, Welzel TM, Zeuzem S, Lawitz EJ, Rodriguez-Torres M, Almeida SS, Resende FF, Ferreira NR, Titze-de-Almeida
Kupcova V, Wiercinska-Drapalo A, Hodges MR, Janssen HL, R, Defino HL, Del Bel E. The Presence of the Neuronal
Reesink HW. Long-term safety and efficacy of microRNA- Nitric Oxide Synthase Isoform in the Intervertebral Disk.
targeted therapy in chronic hepatitis C patients. Antivir Res. Neurotox Res. 2016.
2014;111:53–9. 147. Titze-de-Almeida SS, Lustosa CF, Horst CH, Bel ED,
128. Ottosen S, Parsley TB, Yang L, Zeh K, van Doorn LJ, van der Titze-de-Almeida R. Interferon gamma potentiates the in-
Veer E, Raney AK, Hodges MR, Patick AK. In vitro antiviral jury caused by MPP(+) on SH-SY5Y cells, which is atten-
activity and preclinical and clinical resistance profile of miravirsen, uated by the nitric oxide synthases inhibition. Neurochem
a novel anti-hepatitis C virus therapeutic targeting the human Res. 2014;39(12):2452–64.
factor miR-122. Antimicrob Agents Chemother. 2015;59(1): 148. Felgner PL, Gadek TR, Holm M, Roman R, Chan HW, Wenz
599–608. M, Northrop JP, Ringold GM, Danielsen M. Lipofection: a highly
129. Ojha CR, Rodriguez M, Dever SM, Mukhopadhyay R, El-Hage efficient, lipid-mediated DNA-transfection procedure. Proc Natl
N. Mammalian microRNA: an important modulator of host- Acad Sci U S A. 1987;84(21):7413–7.
pathogen interactions in human viral infections. J Biomed Sci. 149. Rossi JJ. RNAi therapeutics: SNALPing siRNAs in vivo. Gene
2016;23(1):74. Ther. 2006;13(7):583–4.
130. Buxton ILO, Benet LZ. Pharmacokinetics: The Dynamics of 150. Morrissey DV, Lockridge JA, Shaw L, Blanchard K, Jensen K,
Drug Absortion, Distribution, Metabolism, and Elimination. In: Breen W, Hartsough K, Machemer L, Radka S, Jadhav V, Vaish
Goodman and Gilman's The Pharmacological Basis of N, Zinnen S, Vargeese C, Bowman K, Shaffer CS, Jeffs LB, Judge
Therapeutics. 12th Ed.: McGraw-Hill Companies, Inc.; 2011. A, MacLachlan I, Polisky B. Potent and persistent in vivo anti-
HBV activity of chemically modified siRNAs. Nat Biotechnol.
131. Yu ML. Hepatitis C Treatment from Bresponse-guided^ to
2005;23(8):1002–7.
Bresource-guided^ therapy in the transition era from IFN-
151. [Internet] ABC. LNP Delivery Platform. 2016.
containing to IFN-free regimens. J Gastroenterol Hepatol. 2017.
152. Boado RJ. Blood-brain barrier transport of non-viral gene and
doi:10.1111/jgh.13747.
RNAi therapeutics. Pharm Res. 2007;24(9):1772–87.
132. Wilson RC, Doudna JA. Molecular mechanisms of RNA interfer-
153. de Boer AG, Gaillard PJ. Drug targeting to the brain. Annu Rev
ence. Annu Rev Biophys. 2013;42:217–39.
Pharmacol Toxicol. 2007;47:323–55.
133. Shan G. RNA interference as a gene knockdown technique. Int J
154. Lonser RR, Sarntinoranont M, Morrison PF, Oldfield EH.
Biochem Cell Biol. 2010;42(8):1243–51.
Convection-enhanced delivery to the central nervous system. J
134. Wittrup A, Lieberman J. Knocking down disease: a progress re- Neurosurg. 2015;122(3):697–706.
port on siRNA therapeutics. Nat Rev Genet. 2015;16(9):543–52. 155. Cohen ZR, Ramishetti S, Peshes-Yaloz N, Goldsmith M, Wohl A,
135. Leung RK, Whittaker PA. RNA interference: from gene silencing Zibly Z, Peer D. Localized RNAi therapeutics of chemoresistant
to gene-specific therapeutics. Pharmacol Ther. 2005;107(2):222– grade IV glioma using hyaluronan-grafted lipid-based nanoparti-
39. cles. ACS Nano. 2015;9(2):1581–91.
136. Wall NR, Shi Y. Small RNA: can RNA interference be exploited 156. Querbes W, Ge P, Zhang W, Fan Y, Costigan J, Charisse K,
for therapy? Lancet. 2003;362(9393):1401–3. Maier M, Nechev L, Manoharan M, Kotelianski V, Sah DW.
137. de Fougerolles A, Vornlocher HP, Maraganore J, Lieberman J. Direct CNS delivery of siRNA mediates robust silencing in oligo-
Interfering with disease: a progress report on siRNA-based thera- dendrocytes. Oligonucleotides. 2009;19(1):23–9.
peutics. Nat Rev Drug Discov. 2007;6(6):443–53. 157. Haussecker D, Kay MA. RNA interference. Drugging RNAi
138. Sullenger BA, Nair S. From the RNA world to the clinic. Science. Science. 2015;347(6226):1069–70.
2016;352(6292):1417–20. 158. Wu SY, Lopez-Berestein G, Calin GA, Sood AK. RNAi thera-
139. Ozcan G, Ozpolat B, Coleman RL, Sood AK, Lopez-Berestein pies: drugging the undruggable. Sci Transl Med. 2014;6(240):
G. Preclinical and clinical development of siRNA-based therapeu- 240ps247.
tics. Adv Drug Deliv Rev. 2015;87:108–19. 159. Haussecker D. Current issues of RNAi therapeutics delivery and
140. Orom UA, Kauppinen S, Lund AH. LNA-modified oligonucleo- development. J Control Release. 2014;195:49–54.
tides mediate specific inhibition of microRNA function. Gene. 160. Gooding M, Malhotra M, Evans JC, Darcy R, O'Driscoll CM.
2006;372:137–41. Oligonucleotide conjugates - candidates for gene silencing thera-
141. Stein CA, Hansen JB, Lai J, Wu S, Voskresenskiy A, Hog A, peutics. Eur J Pharm Biopharm. 2016;107:321–40.
Worm J, Hedtjarn M, Souleimanian N, Miller P, Soifer HS, 161. Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang
Castanotto D, Benimetskaya L, Orum H, Koch T. Efficient gene Q, Hoekstra M, Kandasamy P, Kel'in AV, Milstein S, Taneja N,
silencing by delivery of locked nucleic acid antisense O'Shea J, Shaikh S, Zhang L, van der Sluis RJ, Jung ME, Akinc A,
Titze-de-Almeida, David and Titze-de-Almeida

Hutabarat R, Kuchimanchi S, Fitzgerald K, Zimmermann T, van 167. Cuellar TL, Barnes D, Nelson C, Tanguay J, Yu SF, Wen
Berkel TJ, Maier MA, Rajeev KG, Manoharan M. Multivalent X, Scales SJ, Gesch J, Davis D, van Brabant SA, Leake D,
N-acetylgalactosamine-conjugated siRNA localizes in hepatocytes Vandlen R, Siebel CW. Systematic evaluation of antibody-
and elicits robust RNAi-mediated gene silencing. J Am Chem Soc. mediated siRNA delivery using an industrial platform of
2014;136(49):16958–61. THIOMAB-siRNA conjugates. Nucleic Acids Res.
162. Zimmermann TS, Karsten V, Chan A, Chiesa J, Boyce M, 2015;43(2):1189–203.
Bettencourt BR, Hutabarat R, Nochur S, Vaishnaw A, Gollob 168. Zhou G, Wilson G, Hebbard L, Duan W, Liddle C, George J,
J. Clinical proof of concept for a novel hepatocyte-targeting Qiao L. Aptamers: a promising chemical antibody for cancer
GalNAc-siRNA conjugate. Mol Ther. 2017;25(1):71–8. therapy. Oncotarget. 2016;7(12):13446–63.
163. [Internet] AP. Alnylam Pharmaceuticals Discontinues Revusiran 169. McNamara 2nd JO, Andrechek ER, Wang Y, Viles KD, Rempel
Development. Available from: http://investors.alnylam. RE, Gilboa E, Sullenger BA, Giangrande PH. Cell type-specific
com/releasedetail.cfm?ReleaseID=992320. delivery of siRNAs with aptamer-siRNA chimeras. Nat
164. Juliano RL. The delivery of therapeutic oligonucleotides. Nucleic Biotechnol. 2006;24(8):1005–15.
Acids Res. 2016;44(14):6518–48. 170. Berezhnoy A, Castro I, Levay A, Malek TR, Gilboa E. Aptamer-
165. Parakh S, Parslow AC, Gan HK, Scott AM. Antibody-mediated targeted inhibition of mTOR in T cells enhances antitumor im-
delivery of therapeutics for cancer therapy. Expert Opinion on munity. J Clin Invest. 2014;124(1):188–97.
Drug Delivery. 2016;13(3):401–19. 171. Thiel WH, Thiel KW, Flenker KS, Bair T, Dupuy AJ,
166. Lorenzer C, Dirin M, Winkler AM, Baumann V, Winkler McNamara 2nd JO, Miller FJ, Giangrande PH. Cell-
J. Going beyond the liver: progress and challenges of internalization SELEX: method for identifying cell-internalizing
targeted delivery of siRNA therapeutics. J Control RNA aptamers for delivering siRNAs to target cells. Methods
Release. 2015;203:1–15. Mol Biol. 2015;1218:187–99.

You might also like