Title of Invention

"METHODS AND COMPOSITIONS TO INHIBIT P2X7 RECEPTOR EXPRESSION"

Abstract Methods and compositions for the downregulation of P2X7 receptor expression or activity are disclosed. Preferred compositions comprise siNA. The methods and compositions are useful in the treatment of diseases characterised by increased P2X7 receptor activity, such as neuronal degeneration, Alzheimer"s disease, inflammatory diseases, and some cancers.
Full Text FIELD OF THE INVENTION
The present invention relates to methods and compositions for the treatment
and/or the prevention of neurona! degeneration or other diseases related to
high levels of expression or activity of P2X7 receptors (P2RX7), In preferred
embodiments, the invention relates to the use of RNAi technology to
downregulate the expression of P2RX7.
Methods and compositions are provided for the treatment of diseases related to
high levels of P2RX7, which include, but are not limited to, neuronal
degeneration, reperfusion or ischemia in stroke or heart attack, Alzheimer's
disease, inflammatory diseases (such as rheumatoid arthritis, osteoarthritis,
asthma, rhinitis, chronic obstructive pulmonary disease (COPD), inflammatory
bowel disease (IBD) such as Crohn's disease), allergies, autoimmune diseases,
cancer (such as leukaemia or non-melanoma skin cancer), skin-related
conditions (such as psoriasis, eczema, alopecia), retinal diseases and treatment
of pain of neuropathic and inflammatory origin.
BACKGROUND OF THE INVENTION
RNAi-as a tool to downregulate gene expression
Gene targeting by homologous recombination is commonly used to determine
gene function in mammals, but this is a costly and time-consuming process.
Alternatively, the functions of many genes can be determined after mRNA
inhibition with ribozyme or antisense technologies. Although successful in some
situations these technologies have been difficult to apply universally. The
advent of siRNA-directed "knockdown" has sparked a revolution in somatic eel!
genetics, allowing the inexpensive and rapid analysis of gene function in
mammals.
Establishing a convenient and reliable method to knock-out gene expression at
the mRNA level has been a recurrent therne in molecular biology over the last
15 years. In efforts to generate loss-of function cells or organisms, various
molecules that included, for example, antlsense sequences, rlbozymes, and
chimeric oligonudeotides have been tested, but the design of such molecules
was based on trial and error, depending on the properties of the target gene.
Moreover, the desired effects were difficult to predict, and often only weak
suppression achieved (Braasch & Corey, 2002).
After the discovery of the phenomenon in plants in the early 1990s, in 1998
Andy Rre and Craig MeHo for the first time demonstrated with the worm
Caenorhabcfitis efegantsthat dsRNA (double-stranded RNA) may specifically and
selectively Inhibit gene expression in an extremely efficient manner (Rre et a/.,
1998). In their experiment; the sequence of the first strand (the so-called sense
RNA) coincides with that of the corresponding region of the target messenger
RNA (mRNA). The second strand (antisense RNA) Is complementary to this
mRNA. The resulting dsRNA turned out to be far more (several orders of
magnitude) efficient than the corresponding single-stranded RNA molecules (in
particular, antlsense RNA). Rre et a/., 1998 named the phenomenon RNAi for
RNA interference. This powerful gene silencing mechanism has been shown to
operate in several species among most phylogenetic phyla.
RNAi begins when an enzyme named DICER encounters dsRNA and chops it
Into pieces called small-interfering RNAs or siRNAs. This protein belongs to the
RNase HI nuclease family. A complex of proteins gathers up these RNA remains
and uses their code as a guide to search out and destroy any RNAs in the cell
with a matching sequence, such as target mRNA (for review see Bosher &
Labouesse, 2000).
The RNAi phenomenon (Akashi etal., 2001) might be summarized as follows:
• Step 1: dsRNA recognition and scanning process.
• Step 2: dsRNA cleavage through RNase III activity and production of siRNAs.
• Step 3: association of the siRNAs and associated factors in RISC complexes,
• Step 4: recognition of the complementary target mRNA.
• Step B: cleavage of the target mRNA in the centre of the region
complementary to the siRNA.
• Step 6: degradation of the target mRNA and recycling of the RISC complex.
In trying to apply the RNAi phenomenon as a technology for gene knockdown,
it was soon realized that mammalian cells have developed various protective
phenomena against viral infections that could impede the use of this approach.
Indeed, the presence of extremely low levels of viral dsRNA triggers an
tofcerferon response, resulting in a global non-specific suppression of translation,
which In turn triggers apoptosis (Williams, 1997, Gil & Esteban, 2000).
In 2000, a first attempt with dsRNA resulted In the specific Inhibition of 3 genes
(MmGFP under the control of the Elongation Factor la, E-cadherin, and c-mos)
in the mouse oocyte and early embryo. Translational arrest, and thus a PKR
response, was not observed as the embryos continued to develop (Wianny &
Zernicka-Goetz, 2000). One year later, research at Ribopharma AG (Kulmbach,
Germany) first demonstrated the functionality of RNAi in mammalian cells.
Using short (20-24 base pairs) dsRNAs - which are called SIRPLEX™- they
specifically switched off genes even in human cells without initiating the acutephase
response. Similar experiments carried out later by other research groups
(Elbashlr etal., 2001; Caplen eta/., 2001) further confirmed these results.
A year later, Paddison et al. (Paddison et al, 2002) tried to use small RNAs
folded in hairpin structures to inhibit the function of specific genes. This work
was inspired by previous studies showing that some genes in Caenorhabditis
elegans naturally regulate other genes through RNAi by coding for hairpinstructured
RNAs. Tested in a variety of normal and cancer human and mouse
cell lines, short hairpin RNAs (shRNAs) are able to silence genes as efficiently as
their siRNA counterparts. Moreover, shRNAs exhibit better reassociation kinetics
in vivo than equivalent duplexes. Even more important, these authors
generated transgenic cell lines engineered to synthesize shRNAs that exhibit a
long-lasting suppressing effect throughout cell divisions (Eurogentec). Recently,
another group of small RNAs (also comprised in the range of 21-25 nt) was
shown to mediate downreguiation of gene expression. These RNAs, known as
small temporally regulated RNAs (stRNAs), have been described in
Caenorhabditis eSegans were they regulate timing of gene expression during
development. It should be noted that stRNAs and siRNAs, despite obvious
similarities, proceed through different modes of action (for review see Banerjee
& Slack, 2002, In contrast with siRNAs, 22 nt long stRNAs downreguiate
expression of target mRNA after transtational initiation without affecting mRNA
integrity. Recent studies Indicate that the two stRNAs first described in
nematodes are the members of a huge family with hundreds of additional
micro-RNAs (miRNAs) existing in metazoarts (Grosshans & Sack, 2002).
Scientists have initially used RNA1 in several systems, including Caenorhabditis
elegans, Drosophila, trypanosomes, and various other invertebrates. Moreover,
using this approach, several groups have recently presented the specific
suppression of protein biosynthesis in different mammalian cell lines -
specifically in HeLa cells - showing that RNAI Is a broadly applicable method for
gene silencing in vitro. Based on these results, RNAi has rapidly become a well
recognized tool for validating (identifying and assigning) gene functions. RNA
interference employing short dsRNA oligonucleotides will, moreover, permit to
decipher the function of genes being only partially sequenced. RNAi will
therefore become inevitable in studies such as:
• Inhibition of gene expression at the post-transcriptional level in eukaryotic
cells, In this context, RNAi is a straight-forward tool to rapidly assess gene
function and reveal null phenotypes.
• Development of the RNAi technology for use in post-implantation embryos.
• The predominant economic significance of RNA interference is established by
its application as a therapeutic principle. As so, RNAi may yield RNA-based
drugs to treat human diseases.
Inhibition of high levels of P2RX7 to prevent disease.
P2X receptors are membrane ion channels that open in response to the binding
of extracellular ATP (North, 2002). They are abundantly distributed, and
functional responses are seen in neurons, glia, epithelia, endothelia, bone,
muscle, and haematopoietic tissues.
The purinergic P2X7 receptors (P2RX7) are ligand-gated cation channels with a
wide distribution that includes cells of the immune and haematopoietic system
(Di Virgllio et al., 2001; North 2002). Two splice forms of P2RX7 corresponding
to GenBank Accession Numbers NMJ302562 and NM_177427 were initially
identified. However, identification of seven variants of human P2RX7 which
result from alternative splicing has recently been reported (Cheewatrakoolpong
et al., 2005).
Activation of P2RX7 by brief exposure to extracellular ATP opens a channel that
allows Ca2"1" and Na+ influx and K+ efflux and that initiates a cascade of
intracellular downstream events. These include the stimulation of phospholipase
D (EI-Moatassim & Dubyak, 1993; Gargett et al, 1996), the activation of
membrane metalloproteases (Jamieson, et al., 1996; Gu et al, 1998; Sluyter &
Wiley, 2002), and the stimulation of intracellular caspases, which eventually
lead to the apoptotic death of the target cell (Ferrari et at, 1999; Humphreys et
al, 2000). P2RX7 activation also leads to extensive membrane blebbing (Virginio
et al., 1999), which is a typical morphological feature of the apoptotic process.
P2RX7 mediates fast excitatory transmission in diverse regions of the brain and
spinal cord (North, 2002). ATP has recently been identified as a potent
transmitter of astrocytic calcium signalling (Cotrina et al., 1998; Guthrie et al,
1999). Astrocytic calcium signalling seems to be a general mechanism by which
astrocytes respond to a variety of stimuli including synaptic activity, transmitter
exposure and traumatic injury (Fields & Stevens-Graham, 2002). By this means,
local astrocytes transmit calcium signals to neurons within their own
geographical mlcrodomaln. This ATP-dependent process of calcium wave
propagation occurs in the brain as well as in the parenchyma of the spinal cord
(Scemes et al.7 2000, Fam et al., 2000), where it may have a role in extending
local injury.
Preliminary observations Indicate that traumatic injury triggers both AT? release
and calcium signalling (Cook & McCleskey, 2002; Neary et al., 2003, Du et al.,
1999). The feet that P2RX7 Is expressed in spinal cord neurons, including motor
neurons (Deuchars et al., 2001), and that P2RX7 is an ATP-gated cation
channel whose activation directly mediates cell death (Di vlrgllio et a/., 1998),
target P2RX7 as good candidates to be inhibited for the prevention of traumatic
injury consequences as well as of chronic trauma. Delivery of P2RX7
antagonists OxATP or PPADS to rats after acute impact injury significantly
improved functional recovery and diminished cell death in the peritraumatic
zone, reducing both the histological extent and functional sequelae of acute
spinal cord injury (Wang et al., 2004).
A postischemic, time-dependent upregulation of the P2X7 receptor-subtype on
neurons and glial cells has also been demonstrated, and suggests a role for this
receptor in the pathophysiology of cerebral ischemia in vivo (Franke et al.,
2004).
Parvathenani et al have shown a remarkable difference in the staining pattern
for P2RX7 in brain slices of a transgenic mice model of Alzheimer's disease (AD)
(Parvathenani et al, 2003). The intense staining for P2RX7 around plaques can
be the result of up-regulation of the P2X7 receptor and/or aggregation of glia
around plaques. The striking association in vivo between P2X7 receptor-positive
cells and plaques in a transgenic mouse model of AD suggests that antagonists
of P2RX7 could have therapeutic utility in treatment of AD by regulating
pathologically activated microglia.
Extracellular ATP has proven to activate multiple downstream signalling events
in a human T-lymphoblastoid cell line (Budagian et al.; 2003). Both P2RX7
rnRNA and protein have been detected in eight of eleven human
haematopoietic cell lines in a non-lineage-specific manner (Zhang et al., 2004).
Further, bone marrow mononuclear cell samples from 69 leukaemia and 9
myelodysplastic syndrome (MDS) patients (out of 87 and 10 patients,
respectively) were P2RX7 positive at mRNA level. Moreover, both positive rates
and relative expression levels were significantly higher in acute myelogenous
leukaemia (AML), acute tyrnpnobtastk: leukaemia (ALL), chronic myelogenous
leukaemia (CML), and MDS groups than in the normal donor group. After one
course of standard Induction therapies, the remission rate in high P2RX7
expression group was tower than that in either the P2RX7 negative group or the
low P2RX7 expression group (Zhang et al., 2004). Expression and function of
P2RX7 have also been associated with the clinical course of patients affected by
chronic lymphocytic leukaemia (CLL) (Cabrini et al., 2005).
Dendritic cells (DC), which are central in the initiation of adaptive immune
responses (Hart, 1997; Stockwln et al., 2000) express P2RX7 (Mutini et al.,
1999; Berchtold et al., 1999; Ferrari et al., 2000). Further, it has been
demonstrated that activation of P2RX7 in DC opens a cation-selective channel
and leads to rapid and near complete shedding of CD23, the low affinity
receptor for IgE (Sluyter & Wiley, 2002), which has an emerging role in chronic
inflammatory diseases Including rheumatoid arthritis (Bonnefy, 1996).
Electrophysiological data and mRIMA analysis of human and mouse pulmonary
epithelia and other epithelial cells indicate that multiple P2XRs are broadly
expressed in these tissues and that they are active on both apical and
basolateral surfaces (Taylor et al,, 1999).
P2RX7 is also expressed on human cutaneous keratinocytes where it has a role
in the signalling system for regulation of proliferation, differentiation and
apoptosis of epidermis (Greig et al., 2003a; Greig et al., 2003b). Further to the
above-mentioned effects, in response to ATP-binding P2RX7 contributes to the
release of the biologically active inflammatory cytokine interleukin IL-1 beta,
following activation of the cells of immune origin in which it is expressed such
as LPS-primed macrophages (Verhoef et al., 2003). Involvement of P2RX7 in
the production of the Inflammatory response of monocytes/macrophages makes
it a good target against cell-mediated autoimmune disorders such as psoriasis.
Expression of P2RX7 has also been detected on Multer glial cells from the
human retina (Pannfcke et al., 2000) as weil as on pericytes of mlcrovessels
isolated from the rat retina, where they regulate the multlceliular functional
organization of the microvascular network (Kawamura et al., 2003). It has
recently been demonstrated that stimulation of P2RX7 by means of agonists
such as benzoylbenzoyl adenosine triphosphate (BzATP) elevates Ca2+ and kills
retinal ganglion cells (Zhang et al., 2005).
Enhanced P2RX7 activity has been detected in human fibroblasts from diabetic
patients, suggesting a possible pathogenetic mechanism for vascular damage in
diabetes (Solini et al., 2004).
Experiments carried out with mice lacking P2RX7 demonstrate that
inflammatory and neuropathic hypersensitivity is completely absent to both
mechanical and thermal stimuli in mutant mice, whilst normal nociceptive
processing is preserved (Chessell et al., 2005). The knockout animals were
unimpaired in their ability to produce mRNA for pro-IL-1 beta, and cytometric
analysts of paw and systemic cytokines from knockout and wild-type animals
following adjuvant insult suggested a selective effect of the gene deletion on
release of IL-lbeta and IL-10, This piece of evidence, together with the fact
that P2RX7 was upregulated in human dorsal root ganglia and injured nerves
obtained from chronic neuropathic pain patients, served to hypothesise that
P2RX7, via regulation of mature IL-1 beta production, plays a role in the
development of pain of neuropathic and inflammatory origin (Chessell et al.,
2005). Drugs which block this target may have the potential to deliver broadspectrum
a nalgesia.
The above-mentioned experimental evidence, therefore, points to inhibition of
P2RX7 as an efficient treatment for diseases such as neuronal degeneration,
reperfuslon or ischemia in stroke or heart attack, Alzheimer's disease,
inflammatory diseases (such as rheumatoid arthritis, osteoarthntis, asthma,
rhinitis, chronic obstructive pulmonary disease (COPD), inflammatory bowel
disease (IB D) such as Crohn's disease), allergies, autoimmune diseases, cancer
(such as le ukaemia, non-metenoma skin cancer), skin-related conditions (such
as psoriasis, eczema, atopeda), retinal diseases and treatment of pain of
neuropathic and inflammatory origin.
A novel approach to exert this inhibition is the down regulation of P2RX7 gene
expression mediated by RNA interference (RNAi).
BRIEF SUMMARY OF THE INVENTION
In the present invention we describe a method for the treatment and/or
prevention of neuronal degeneration or other diseases related to high levels of
P2RX7. The method is based on the downregulation of expression of one or
more splice forms of the P2RX7 gene. Downregulation may be effected by the
use of double stranded nucleic acid moieties, named siNA or small interfering
NA that are directed at interfering with the mRNA expression of either one or
more splicing forms of the P2RX7 gene. The silMA are preferably siRNA,
although modified nucleic acids or similar chemically synthesised entities are
also i nduded within the scope of the invention.
Embodiments of the invention provide pharmaceutical compositions for use in
the treatment of neuronal degeneration conditions and of other animal
(including human) diseases related to high levels of P2RX7.
DETAILED DESCRIPTION OF THE INVENTION
Design of siNA
Although the mechanisms for RNAi remain unknown, the steps required to
generate the specific dsRNA oligonudeotides are dear. It has been shown that
dsRNA duplex strands that are 21-26 nucleotfdes in length work most
effectively in produdng RNA interference. Selecting the right homologous
region within the gene is also important. Factors such as the distance from start
codon, the G/C content and the location of adenosine dimers are important
when considering the generation of dsRNA for RNA5. One consequence of this,
however, is that one may need to test several different sequences for the most
efficient RNAi and this may become costly.
In 1999, Tuschl et al. deciphered the silencing effect of siRIMAs showing that
their efficiency is a function of the length of the duplex, the length of the 3'-end
overhangs, and the sequence in these overhangs. Based on this founder work,
Eurogentec recommends that the target mRNA region, and hence the sequence
of the siRNA duplex, should be chosen using the following guidelines:
Since RNAi relies on the establishment of complex protein interactions, it is
obvious that the mRNA target should be devoided of unrelated bound factors.
In this context, both the 5' and 3' untranslated regions (UTRs) and regions
close to the start codon should be avoided as they may be richer in regulatory
protein binding sites. The sequence of the siRNA is therefore selected as
follows:
. In the mRNA sequence, a region located 50 to 100 nt downstream of the
AUG start codon or upstream of stop codon is selected.
. In this region, the following sequences are searched for: M(N19), CA(N19).
. The G/C percentage for each identified sequence is calculated. Ideally, the
G/C content is 50 % but it must less than 70 % and greater than 30 %.
Preferably, sequences containing following repetitions are avoided: AAA,
CCC, GGG, TTT, AAAA, CCCC, GGGG, TTTT.
• An accessibility prediction according to the secondary structure of the
mRNA is carried out as well.
• A BLAST is also performed (i.e. NCBI ESTs database) with the nucleotide
sequence fitting best the previous criteria to ensure that only one gene will
be inactivated.
In order to maximize the result's interpretation, the following precautions
should be taken when using sJRNAs:
• Always test the sense and antisense single strands in separate experiments.
• Try a scramble siRNA duplex. This should have the same nucleotide
composition as your siRNA but lack significant sequence homology to any other
gene (including yours).
• If possible, knock-down the same gene with two independent siRNA duplexes
to control the specificity of the silencing process.
Practically, each of the selected genes is introduced as a nucleotide sequence in
a prediction program that takes into account all the variables described above
for the design of optimal ollgonucleotides. This program scans any mRNA
nucleotide sequence for regions susceptible to be targeted by siRNAs. The
output of this analysis is a score of possible siRNA oligonucleotides. The highest
scores are used to design double stranded RNA oligonucleotides (typically 21 bp
long, although other lengths are also possible) that are typically made by
chemical synthesis.
In addition to siRNA, modified nucleotides may also be used. We plan to test
several chemical modifications that are well known in the art. These
modifications are aimed at increasing stability or availability of the siNA.
Examples of suitable modifications are described in the publications referenced
below, each of which is incorporated herein by reference.
Studies show that replacing the B'-terminal nucleotide overhanging segments of
a 21-mer siRNA duplex having two -nucleotide 3'-overhangs with
deoxyribonucleotides does not have an adverse effect on RNAi activity.
Replacing up to four nucleotides on each end of the siRNA with
deoxyribonucleotides has been reported to be well tolerated, whereas complete
substitution with deoxyribonucleotides results in no RNAi activity (Elbashir
2001). In add-on, Boashir et al. also report that substrtutjon of siRNA with 21-
O methyl nudeotides completely abolishes RNAi activity.
Affinity modified nudeosides as described in WO2005/044976 may be used.
This publication describes oligonudeotides comprising nudeosides modified so
as to have increased or decreased affinity for their complementary nucleotide in
the target mRNA and/or in the complementary siNA strand.
GB2406568 describes alternative modified oligonudeotides chemically modified
to provide improved resistance to degradation or improved uptake. Examples of
such modifications include phosphorothioate internucleotide linkages, 2'-0-
methyl ribonucleotides, 2-deoxy-fluoro ribonucleotides, 2'-deoxy
ribonucleotides, "universal base" nucleotides, 5-C-methyl nucleotides, and
inverted deoxyabasic residue incorporation.
WO2004/029212 describes oligonudeotides modified to enhance the stability of
the siRNA or to increase targeting efficiency. Modifications include chemical
cross linking between the two complementary strands of an siRNA and chemical
modification of a 3' terminus of a strand of an siRNA. Preferred modifications
are internal modifications, for example, sugar modifications, nucleobase
modifications and/or backbone modifications. 2'-fluoro modified ribonucleotides
and Z'-deoxy ribonucleotides are described.
W02005/040537 further recites modified oligonucleotides which may be used in
the invention.
As weJI as making use of dsNA and modified dsNA, the present invention may
use short hairpin NA (shNA); the two strands of the siNA molecule may be
connected by a linker region, which may be a nucleotide linker or a nonnudeotide
linker.
In addition to siNA which is perfectly complementary to the target region,
degenerate siNA sequences may be used to target homologous regions.
WO2005/045037 describes the design of siNA molecules to target such
homologous sequences, for example by incorporating non-canonical base pairs,
for example mismatches and/or wobble base pairs, that can provide additional
target sequences. In instances where mismatches are identified, non-canonical
base pairs (for example, mismatches and/or wobble bases) can be used to
generate siNA molecules that target more than one gene sequence. In a nonlimiting
example, non-canonical base pairs such as UU and CC base pairs are
used to generate siNA molecules that are capable of targeting sequences for
differing targets that share sequence homology. As such, one advantage of
using siNAs of the invention is that a single siNA can be designed to include
nucleic acid sequence that is complementary to the nucleotide sequence that is
conserved between homologous genes. In this approach, a single siNA can be
used to inhibit expression of more than one gene instead of using more than
one siNA molecule to target different genes.
Preferred siNA molecules of the Invention are double stranded. A siNA molecule
of the invention may comprise blunt ends, that is, ends that do not include any
overhanging nudeotides. In one embodiment, an siNA molecule of the
invention can comprise one or more blunt ends. In preferred embodiments, the
siNA molecules have 3' overhangs. siNA molecules of the invention may
comprise duplex nucleic acid molecules with 3' overhangs of n nudeotides
(5>n>l). Elbashir (2001) shows that 21-nucleotide siRNA duplexes are most
active when containing 3'-terminal dinucleotide overhangs.
Candidate oligonucleotides are further filtered for interspecles sequence
conservation in order to facilitate the transition from animal to human clinical
studies. In preferred embodiments of the invention, conserved oligonucleotides
are used; this allows a single oligonudeotide sequence to be used in both
animal models and human dinical trials.
GenBank Accession Numbers corresponding ID P2RX7 transcripts produced by
alternative spitoing are displayed in Figure 1.
Selected oligonudeotide sequences against which RNAi is directed are shown in
Rgure 2. Displayed sequences are the DNA sequences targeted by the siNA.
Therefore, the invention would make use of NA duplexes with sequences
complementary to the indicated DNA sequences.
The sequences displayed in Figure 2 are not limiting. As a matter of fact, target
DNA need not necessarily be preceded by AA or CA. Further, target DNA could
be constituted by sequences included in Figure 2 flanked by any contiguous
sequence.
In vitro studies.
Obtaining siRNA duplexes
RNAs are preferably chemically synthesized using appropriately protected
ribonucleoside phosphoramidites and a conventional DNA/RNA synthesizer.
Substitution of one or both strands of a siRNA duplex by 2'-deoxy or 2'-O
methyl oligoribonucleotides abolished silencing in fly extract (Elbashir et al.
2001). In mammalian cells, however, it seems possible to substitute the sense
siRNA by a 2'-O-methyl oligoribonucleotide (Ge et al. 2003).
Most conveniently, siRNAs are obtained from commercial RNA oligo synthesis
suppliers, which sell RNA-synthesis products of different quality and costs. In
general, 21-nt RNAs are not too difficult to synthesize and are readily provided
in a quality suitable for RNAI.
Suppliers of RNA synthesis reagents include Proligo (Hamburg, Germany),
Dharmacon Research (Lafayette, CO, USA), Glen Research (Sterling, VA, USA),
ChemGenes (Ashland, MA, USA), and Cruachem (Glasgow, UK), Qiagen
(Germany), Ambton (USA) and Invitrogen (Scotland). The previous custom RNA
synthesis companies are entitled to provide siRNAs with a license for target
validation. In particular, our siRNA suppliers are Amblon, Dharmacon and
Invitrogen, companies that offer the traditional custom chemical synthesis
service for siRNA, and supply the siRNA with HPLC purification and delivered in
dry form along with RNase-free water. A central web-based resource for RNAi
and siRNA methodologies, along with links to additional siRNA related products
and services, can be found on the website of above-mentioned suppliers.
An annealing step is necessary when working with single-stranded RNA
molecules. It is critical that all handling steps be conducted under sterile, Rnase
free conditions. To anneal the RNAs, the oligos must first be quantified by UV
absorption at 260 nanometres (nm). The following protocol based on Elbashir
et al. (2001) is then used for annealing:
• Separately aliquot and dilute each RNA oligo to a concentration of 50 uM.
• Combine 30 ul of each RNA oligo solution and 15 ul of 5X annealing buffer.
Final buffer concentration is: 100 mM potassium acetate, 30 mM HEPES-KOH
pH 7.4, 2 mM magnesium acetate. Final volume is 75 ul.
• Incubate the solution for 1 minute at 90 °C, centrifuge the tube for 15
seconds, let sit for 1 hour at 37 °C, then use at ambient temperature. The
solution can be stored frozen at -20 °C and freeze-thawed up to 5 times. The
final concentration of siRNA duplex is usually 20 uM.
Alternatively, already annealed dsRNAs may be purchased from the suppliers.
Chemically modified nucleic acids may also be used. For example, an overview
of the types of modification which may be used is given in WO03/070744, the
contents of which are incorporated herein by reference. Particular attention is
drawn to pages 11 to 21 of this publication. Other possible modifications are as
described above. The skilled person will be aware of other types of chemical
modification which may be incorporated into RNA molecules.
*In vitro" system
To check the spedflcHy of the sIRNA interference cell cultures and organotypic
cultures both expressing the target gene, were employed.
The cells used for these experiments were murine muscle cells, .C2C12, and the
organotypic cultures were spinal cord slices. The levels of P2RX7 expression
were analyzed after being incubated with the corresponding siRNA duplexes.
For linking siRNA knockdown to specific phenotypes in cultured cells, it is
necessary to demonstrate the decrease of the targeted protein or at least to
demonstrate the reduction of the targeted mRNA.
mRNA levels of the target gene can be quantitated by Real-time quantitative
PCR (qRT-PCR). Further, the protein levels can be determined in a variety of
ways well known in the art, such as Western blot analysis with specific
antibodies to the different target allow direct monitoring of the reduction of
targeted protein.
Transfection of siRNA duplexes in cell cultures.
Several examples of techniques well known in the art are as follows: We can
perform a single transfection of siRNA duplex using a cationic lipid, such as
Lipofectamine 2000 Reagent (Invitrogen) and assay for silencing 24, 48 and 72
hours after transfection.
A typical transfection protocol can be performed as follows: For one well of a 6-
well plate, we transfect using 100 or 200nM as final concentration of siRNA.
Following Lipofectamine 2000 Reagent protocol, the day before transfection, we
seed 2-4 x 10s cells per well in 3ml of an appropriate growth medium,
containing DMEM, 10% serum, antibiotics and glutamine, and incubate cells
under normal growth conditions (37°C and 5% CO2). On the day of
transfection, celte have to be at 30-50% confluence. We dilute 12.5ul of 20uM
siRNA duplex (corresponding to 100 nM final concentration) or 25ul of 20uM
siRNA duplex (corresponding to 200nM final concentration) in 250ul of DMEM
and mix. Also, 6ul of Lipofectamine 2000 is diluted in 250ul of DMEM and
mixed. After a 5 minutes incubation at room temperature, the diluted oligomer
(siRNA duplex) and the diluted Upofectamine are combined to allow complex
formation during a 20 minutes incubation at room temperature. Afterwards, we
add the complexes drop-wise onto the cells with 2 ml of fresh growth medium
low in antibiotics and mix gently by rocking the plate back and forth, to ensure
uniform distribution of the transfection complexes. We incubate the cells under
their normal growth conditions and the day after the complexes are removed
and fresh and complete growth medium is added. To monitor gene silencing
cells are collected at 24,48 and 72h post-transfection.
The efficiency of transfection may depend on the cell type, but also on the
passage number and the confluency of the cells. The time and the manner of
formation of siRNA-liposome complexes (e.g. inversion versus vortexing) are
also critical. Low transfection efficiencies are the most frequent cause of
unsuccessful silencing. Good transfection is a non-trivial issue and needs to be
carefully examined for each new cell line to be used. Transfection efficiency
may be tested transfecting reporter genes, for example a CMV-driven EGFPexpression
plasmid (e.g. from Clontech) -or a B-Gal expression plasmid, and
then assessed by phase contrast and/or fluorescence microscopy the next day.
Testing of siRNA duplexes
Depending on the abundance and the life time (or turnover) of the targeted
protein, a knock-down phenotype may become apparent after 1 to 3 days, or
even later. In cases where no phenotype is observed, depletion of the protein
may be observed by immunofiuorescence or Western blotting.
After transfections, total RNA fractions extracted from cells were pre-treated
with DNase I and used for reverse transcription using a random primer. PCRamplifled
with a specific primer pair covering at least one exon-exon junction in
order to control for amplification of pre-mRNAs. RT/PCR of a non-targeted
mRNA is also needed as control. Effective depletion of the mRNA yet
undetectabte reduction of target protein may indicate that a large reservoir of
stable protein may exist in the cell. Alternatively, Real-time PCR amplification
can be used to test in a more precise way the mRNA decrease or
disappearance. Real-time reverse-transcriptase (RT) PCR quantitates the Initial
amount of the template most specifically, sensitively and reproducibly. Realtime
PCR monitors the fluorescence emitted during the reaction as an indicator
of amplicon production during each PCR cycle, in a light cycler apparatus. This
signal increases in direct proportion to the amount of PCR product in a reaction.
By recording the amount of fluorescence emission at each cycle, it is possible to
monitor the PCR reaction during exponential phase where the first significant
increase in the amount of PCR product correlates to the initial amount of target
template.
To verify the interference pattern of the differentially expressed P2RX7 gene in
the cell cultures, qRT-PCR was performed according to the manufacturer
protocol (Roche). For quantitative qRT-PCR, approximately 500 ng of total RNA
were used for reverse transcription followed by PCR amplification with specific
primers for each gene in reaction mixture containing Master 5YBR Green I. The
PCR conditions were an initial step of 30 min at 91 °C, followed by 40 cycles of 5
s at 95°C, 10 s at 62°C and 15 s at 72°C. Quantification of b-actin mRNA was
used as a control for data normalization. Relative gene expression comparisons
work best when the gene expression of the chosen endogenous/internal control
is more abundant and remains constant, in proportion to total RNA, among the
samples. By using an invariant endogenous control as an active reference,
quantltation of an mRNA target can be normalised for differences in the amount
of total RNA added to each reaction. The amplification curves obtained with the
light cycler were analyzed in combination with the control kit RNA, which
targets in vitro transcribed cytokine RNA template, according to the
manufacturer protocol. In order to assess the specificity of the amplified PCR
product a melting curve analysis was performed. The resulting melting curves
allow discrimination between primer-dimers and specific PCR product.
Transfectkm of siRNA duplexes in organotypic cultures
To obtain spinal cord organotypic cultures, the experimental protocol was
performed as follows: The spinal cord was extracted from 6 to 8 week old rats
and placed in ice-cold dissecting media containing Gey's Medium supplemented
with D-Glucose (6.5 mg/ml) and 15mM Hepes. To generate the organotypic
cultures, 500 urn slices from the thoracic spinal cord were obtained using a
tissue chopper and placed in sterile MEM supplemented with Earl's salt solution.
Spinal slices were transferred onto Millicell culture plates. Each plate, containing
4 to 6 slices, was placed into wells of a six-well plate containing 1.25ml of
antibiotic-free medium (50% MEM with Earl's salts and glutamine, 25% Hanks
balanced salt solution and 25% Horse Serum supplemented with D-Glucose
(6mg/ml) and 20mM Hepes).
Slices were incubated under normal growth conditions (37°C and 5% C02) and
media was changed the day after and, afterwards, three times a week.
In these conditions, spinal cord organotypic cultures maintain their structural
integrity for at least 15 days and present high levels of the P2RX7 transcript.
Also, P2RX7 gene expression, checked by quantitative PCR assays, does not
present any relevant change along the culture period.
To perform siRNA transfections there are several protocols and techniques well
known in the art. In this case a double transfection is needed to observe an
enhanced gene expression inhibition. Double siRNA transfections were
performed using a cationic lipid, such as Lipofectamine 2000 Reagent
(Invitrogen) and gene expression silencing was assayed at different time points.
A typical transfection protocol can be performed as folbws. Each Millicell culture
plate, containing 4 to 6 slices, is transfected using a determined concentration
of siRNA. Following Upofectamlne 2000 Reagent protocol for siRNA
transfection, we dilute the amount of siRNA duplex in 50ul of MEM and mix. In
a different tube, the Lipofectamine 2000 are diluted in 50ul of MEM and mixed.
After 5 minutes incubation at room temperature, the diluted siRNA and the
diluted Lipofectamine are combined to allow complex formation during 20
minutes incubation at room temperature. Afterwards, the complexes are added
drop-wise over the slices. We incubate the slices under their normal growth
conditions and the day after, the complexes are removed and fresh and
complete growth medium is added. When necessary, 48h after the first
Lipofectamine treatment the protocol is repeated as previously described.
The efficiency of transfection may depend on the cell or tissue type, but also on
their culture characteristics. The time and the manner of formation of siRNAliposome
complexes are also critical. Low transfection efficiencies are the most
frequent cause of unsuccessful silencing. Good transfection is a non-trivial issue
and needs to be carefully examined for each new cell line to be used.
Transfection efficiency may be tested transfecting reporter genes, for example
a CMV-driven EGFP-expression plasmid (e.g. from Clontech) or a p-Gal
expression plasmid, and then assessed by phase contrast and/or fluorescence
microscopy the next day. Spinal cord organotypic cultures were successfully
transfected with a p-Gal encoded construct reporter. The enzymatic activity of
bacterial. p-Gal can be assayed readily from transfected tissue with an
appropriate commercial staining set.
Pharmaceutical formulations
The present invention may comprise the administration of one or more species
of siNA molecule simultaneously. These species may be selected to target one
or more target genes.
The SJNA molecules of the Invention and formulatjons or compositions thereof
may be administered directly or topically (e. g., locally) to the organ of interest
(for example, spinal cordf brain, etc) as is generally known in the art For
example, a sINA molecule can comprise a delivery vehicle, including liposomes,
for administration to a subject. Carriers and diluents and their salts can be
present in pharmaceutlcally acceptable formulations. Nucleic acid molecules can
be administered to cells by a variety of methods known to those of skill in the
art, including, but not restricted to, encapsulation in liposomes, by
iontophoresis, or by incorporation into other vehicles, such as biodegradable
polymers, hydrogels, cyclodextrins poly (lactic-co-glycolic) acid (PLGA) and
PICA microspheres, biodegradable nanocapsuies, and bioadhesive
microspheres, or by proteinaceous vectors. In another embodiment, the nucleic
acid molecules of the invention can also be formulated or complexed with
polyethyleneimine and derivatives thereof, such as polyethyleneiminepolyethyleneglycol-
N-acetylgalactosamine (PEI-PEG-GAL) or polyethyleneiminepolyethyleneglycol-
tri-N-acetylgalactosamine (PEI-PEG-triGAL) derivatives.
A siNA molecule of the invention may be complexed with membrane disruptive
agents and/or a cationic lipid or helper lipid molecule.
Delivery systems which may be used with the invention include, for example,
aqueous and non aqueous gels, creams, multiple emulsions, microemulsions,
liposomes, ointments, aqueous and non aqueous solutions, lotions, aerosols,
hydrocarbon bases and powders, and can contain excipients such as
solubilizers, permeation enhancers (e. g., fatty acids, fatty acid esters, fatty
alcohols and amino acids), and hydrophilic polymers (e. g. , polycarbophil and
polyvinylpyrolidone). In one embodiment, the pharmaceutically acceptable
carrier is a liposome or a transdermal enhancer.
A pharmaceutical formulation of the invention is in a form suitable for
administration, e.g., systemic or local administration, into a cell or subject,
Including for example a human. Suitable forms, in part; depend upon the use or
the route of entry, for example oral, transdermal, or by injection. Other factors
are known in the art, and include considerations such as toxidty and forms that
prevent the composition or formulation from exerting its effect.
The present invention also includes compositions prepared for storage or
administration that include a pharmaceuticaHy effective amount of the desired
compounds in a pharmaceutically acceptable carrier or diluent. Acceptable
carriers or diluents for therapeutic use are well known in the pharmaceutical
art. For example, preservatives, stabilizers, dyes and flavouring agents can be
provided. These include sodium benzoate, sorbic acid and esters of phydroxybenzoic
acid. In addition, antioxidants and suspending agents can be
used.
A pharmaceutically effective dose is that dose required to prevent, inhibit the
occurrence, or treat (alleviate a symptom to some extent, preferably all of the
symptoms) of a disease state. The pharmaceutically effective dose depends on
the type of disease, the composition used, the route of administration, the type
of mammal being treated, the physical characteristics of the specific mammal
under consideration, concurrent medication, and other factors that those skilled
in the medical arts will recognize.
Generally, an amount between O.lmg/kg and 100 mg/kg body weight/day of
active ingredients is administered.
The formulations of the invention can be administered in unit dosage
formulations containing conventional non-toxic pharmaceutically acceptable
carriers, adjuvants and/or vehicles. Formulations can be in a form suitable for
oral use, for example, as tablets, troches, lozenges, aqueous or oily
suspensions, dlspersible powders or granules, emulsion, hard or soft capsules,
or syrups or elixirs. Compositions intended for oral use can be prepared
according to any method known to the art for the manufacture of
pharmaceutical compositions and such compositions can contain one or more
such sweetening agents, flavouring agents, colouring agents or preservative
agents in order to provide pharmaceutkally eiegant and palatable preparations.
Tablets contain the active ingredient in admixture with non-toxic
pharmaceutically acceptable exdpients that are suitable for the manufacture of
tablets.
These excipients can be, for example, inert diluents; such as calcium carbonate,
sodium carbonate, lactose, calcium phosphate or sodium phosphate;
granulating and disintegrating agents, for example, corn starch, or alginlc acid;
binding agents, for example starch, gelatin or acacia; and lubricating agents,
for example magnesium stearate, stearic acid or talc. The tablets can be
uncoated or they can be coated by known techniques. In some cases such
coatings can be prepared by known techniques to delay disintegration and
absorption in the gastrointestinal tract and thereby provide a sustained action
over a longer period. For example, a time delay material such as glyceryl
monostearate or glyceryl distearate can be employed.
Formulations for oral use can also be presented as hard gelatin capsules
wherein the active ingredient is mixed with an inert solid diluent, for example,
calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules
wherein the active ingredient is mixed with water or an oil medium, for example
peanut oil, liquid paraffin or olive oil.
Aqueous suspensions contain the active materials in a mixture with excipients
suitable for the manufacture of aqueous suspensions. Such excipients are
suspending agents, for example sodium carboxymethylcellulose,
methylcellulose, hydropropyl- methylcellufose, sodium alginate,
polyvinylpyrrondone, gum tragacanth and gum acacia; dispersing or wetting
agents can be a naturally-occurring phosphatide, for example, lecithin, or
condensation products of an atkytene oxide with fatty adds, for example
poJyoxyethylene stearate, or condensation products of ethylene oxide with long
chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or
condensation products of ethytene oxide with partial esters derived from fatty
acids and a hexitol such as polyoxyethyiene sorbitol monooleate, or
condensation products of ethylene oxide with partial esters derived from fatty
acids and hexitol anhydrides, for example polyethylene sorbitan monooleate.
The aqueous suspensions can also contain one or more preservatives, for
example ethyl, or n-propyl p-hydroxybenzoate, one or more colouring agents,
one or more flavouring agents, and one or more sweetening agents, such as
sucrose or saccharin.
Oily suspensions can be formulated by suspending the active ingredients in a
vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a
mineral oil such as liquid paraffin. The oily suspensions can contain a thickening
agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents
and flavouring agents can be added to provide palatable oral preparations.
These compositions can be preserved by the addition of an anti-oxidant such as
ascorbic acid.
Dispersible powders and granules suitable for preparation of an aqueous
suspension by the addition of water provide the active ingredient in admixture
with a dispersing or wetting agent, suspending agent and one or more
preservatives. Suitable dispersing or wetting agents or suspending agents are
exemplified by those already mentioned above. Additional excipients, for
example sweetening, flavouring and colouring agents, can also be present.
Pharmaceutical compositions of the invention can also be in the form of oil-inwater
emulsions. The oily phase can be a vegetable oil or a mineral oil or
mixtures of these. Suitable emulsifying agents can be naturally-occurring gums,
for example gum acacia or gum tragacanth, naturally-occurring phosphatides,
for example soy bean, lecithin, and esters or partial esters derived from fatty
adds and hexitol, anhydrides, for example sorbitan monooleate, and
condensation products of the said partial esters with ethylene oxide, for
example polyoxyethytene sorbitan monooleate. The emulsions can also contain
sweetening and flavouring agents.
Syrups and elixirs can be formulated with sweetening agents, for example
glycerol, propylene glycol, sorbitol, glucose or sucrose. Such formulations can
also contain a demulcent, a preservative and flavouring and colouring agents.
The pharmaceutical compositions can be in the form of a sterile injectable
aqueous or oleaginous suspension.
This suspension can be formulated according to the known art using those
suitable dispersing or wetting agents and suspending agents that have been
mentioned above.
A sterile injectable preparation can also be a sterile injectable solution or
suspension in a non-toxic parentally acceptable diluent or solvent, for example
as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents
that can be employed are water, Ringer's solution and isotonic sodium chloride
solution. In addition, sterile, fixed oils are conventionally employed as a solvent
or suspending medium. For this purpose, any bland fixed oil can be employed
including synthetic mono-or diglycerides. In addition, fatty acids such as oleic
acid find use in the preparation of injectables.
The nucleic acid molecules of the invention can also be administered in the
form of suppositories, e. g. , for rectal administration of the drug. These
compositions can be prepared by mixing the drug with a suitable non-irritating
excipient that is solid at ordinary temperatures but liquid at the rectal
temperature and will therefore melt in the rectum to release the drug. Such
materials include cocoa butter and polyethylene glycols.
Nudeic acid molecules of the invention can be administered parenterally in a
sterile medium. The drug, depending on the vehicle and concentration used,
can either be suspended or dissolved in the vehicle. Advantageously, adjuvants
such as local anaesthetics, preservatives and buffering agents can be dissolved
in the vehicle.
It is understood that the specific dose level for any particular subject depends
upon a variety of factors including the activity of the specific compound
employed, the age, body weight, general health, sex, diet, time of
administration, route of administration, and rate of excretion, drug combination
and the severity of the particular disease undergoing therapy.
For administration to non-human animals, the composition can also be added to
the animal feed or drinking water. It can be convenient to formulate the animal
feed and drinking water compositions so that the animal takes in a
therapeutically appropriate quantity of the composition along with its diet. It
can also be convenient to present the composition as a premix for addition to
the feed or drinking water.
The nucleic acid molecules of the present invention can also be administered to
a subject in combination with other therapeutic compounds to increase the
overall therapeutic effect The use of multiple compounds to treat an indication
ca n increase the beneficial effects while reducing the presence of side effects.
Alternatively, certain siNA molecules of the invention can be expressed within
cells from eukaryotic promoters. Recombinant vectors capable of expressing the
siNA molecules can be delivered and persist in target cells. Alternatively, vectors
can be used that provide for transient expression of nucleic acid molecules.
Such vectors can be repeatedly administered as necessary. Once expressed, the
siNA molecule interacts with the target mRNA and generates an RNAi response.
Delivery of siNA motecute expressing vectors can be systemic, such as by
intravenous or Intra-rnuscular administration, by administration to target cells
ex-planted from a subject followed by relntroduciion Into the subject, or by any
other means that would allow for Introduction into the desired target cell.
Results
Example 1. In vitro assays.
A panel of siRNA against the P2RX7 target gene has been analyzed. The first
step was to perform experiments in cell cultures. For the P2RX7 target gene,
several siRNAs were designed using a specific software according to the rules
described before. Those with the best characteristics were selected to be
tested. The siRNAs were applied to cell cultures, such as C2C12. The effect of
siRNAs over the target gene was analyzed by Real-time PCR according to the
manufacturer's protocol. The gene target transcript levels were normalized
using actin as housekeeping gene. Some of the different siRNAs that were
tested and their different efficacies in the interference of the target gene are
included in Rgure 3. RNA was prepared from C2C12 cells treated with different
siRJMAs for 48h. The samples were analyzed by real time PCR using specific
primers. The values show the mean expression levels of different transcripts
normalized to actin relative to cell control. siRNAl, siRNA2 and siRNA3 target
the murine sequences homologous to the human sequences listed in Figure 2
as follows: siRNAl targets the murine sequence homologous to human SEQ, ID.
37; siRNAZ targets the murine sequence homologous to human SEQ. ID. 78;
and SIRNA3 targets the murine sequence homologous to human SEQ. ID. 92.
The values represent the mean of the percentage of the normalized mRNA
levels upon siRNA interference over the control gene expression and their
standard deviations. The level of the P2RX7 transcript after the siRNA treatment
was highly reduced with siRNA2 and siRNA3, compared to the control cells. The
decrease of the gene expression depends on the efficiency in siRNA silencing.
In fact, siRNA2 treatment decreased the P2RX7 gene expression to 58 %
compared to the control.
Example 2. Time-dose response in vitro.
In order to validate the efficiency of siRNAZ, more treatments were carried out
in C2C12 cells. Cells were transfected with sfRNAZ and the level of P2RX7
transcript was analyzed by Real-time PCR at 24, 48 and 72 h. The level of the
transcript was significantly reduced at this time points after the siRNA
treatment. Rgure 4 shows the mean of the percentage of the normalized mRNA
P2RX7 levels upon siRNA interference over the control gene expression at each
time point and their standard deviations. Moreover a siRNA dose-response was
analyzed. Figure 4 shows the results of two different siRNA applications (100
and 200nm). 200nm siRNA applications were more effective in the P2RX7
downregulatlon than those with lOOnm, confirming both the specificity and the
effectiveness of the treatment.
Example 3. Organotypic cultures.
Previously to the siRNA spinal cord application, we performed in vitro
experiments using an established model based on spinal cord slice cultures. It is
essential to use appropriate experimental models in order to understand the
complex processes which evolve after the initial trauma. This model facilitated
the investigation of primary and secondary mechanisms of cell death that
occurs after spinal cord injury and represents a step before study of the effect
of P2RX7 interference in murine models is undertaken. Several in vivo models
have been characterized to study the chronic pathology. However, due to the
complexity of the in vivo system, interpretation of results may be more difficult,
plus the cost of maintaining an animal model is very high. In order to study the
events after trauma the in vitro models are preferred, as these allow precise
control over the environment, and easy and repeated access.
Previously to the experiment, the morphological integrity of cultures was
examined by microscopy and the delivery in this model was carried out
transfectrng a reporter gene (p-gal) according to the described protocol. Upon
transfecting cells with a p-gal construct, spinal cords were fixed, washed and
stained with freshly prepared X-gal staining solution. Blue staining appeared
upon 24 h indicating a successfully delivery.
Two siRNAs were selected according to different criteria, siRNAH and siRNAR.
siRNAH targets the rat sequence homologous to human SEQ. ID. 58 of Figure
2, while siRNAR targets the best candidate sequence in rat, selected by specific
software, homologous to human SEQ. ID. 37 of Figure 2. Spinal cord cultures
were obtained as previously described. Each plate was double transfected with
the corresponding siRNA and gene expression was assayed at 72 and 96 h after
the first transfection by Real- time PCR.
Figure 5 shows a representative experiment. Values represent the percentage
of mRNA transcript relative to the control after siRNA treatment once
normalized using IBS as the reference housekeeping gene. Previous
experiments indicated 18S as a better reference than p actin in spinal cord
cultures analysis. The decrease in P2RX7 transcript is higher at 72 h being
around 70%, at 96h there is less reduction, around 80-90%. This set of
experiments confirms the ability of RNAi to reduce P2RX7 expression in an
organotypic culture very close to an in vivo model.
REFERENCES
Akashi H, Miyagishi M, Taira K. Suppression of gene expression by RNA
interference in cultured plant cells. Antisense Nucleic Acid Drug Dev, 2001,
11(6)1359-67.
Banerjee D, Slack F. Control of developmental timing by small temporal RNAs: a
paradigm for RNA-mediated regulation of gene expression. Bioessays, 2002,
24(2): 119-29.
Berchtold, S., Ogilvie, A. L 3., Bogdan, C, Muhl-Zurbes, P., Ogilvie, A., Schuler,
G. and Steinkasserer, A. Human monocyte derived dendritic cells express
functional P2X and P2Y receptors as well as ecto-nudeotidases. FEBS Lett.,
1999, 458:424.
Bonnefy, 1-Y., Pteter-Zyberk, C, Lecoanet-Henchoz, S., Gauchat, J.-F., Aubry,
J.-P. and Graber, P. A new role for CD23 in Infammation. Immunol. Today,
1996,17:418.
Bosher JM, Labouesse M. RNA interference: genetic wand and genetic
watchdog. Nat Cell Btol, 2000, 2(2):E31-6.
Braasch DA, Corey DR. Novel antisense and peptide nucleic acid .strategies for
controlling gene expression. Biochemistry, 2002, 41(14):4503-10.
Budagian V, Bulanova E, Brovko L, Orinska Z, Fayad R, Paus R, Bulfone-Paus S.
Signaling through P2X7 receptor in human T cells involves p561ck, MAP kinases,
and transcription factors AP-1 and NF-kappa B. ;i Biol Chem., 2003 Jan 17;
278(3): 1549-60.
Cabrini G, Falzoni S, Forchap SL, Pellegatti P, Balboni A, Agostini P, Cuneo A,
Castoldi G, Baricordi OR, Di Virgilio F. A His-155 to Tyr polymorphism confers
gain-of-function to the human P2X7 receptor of human leukemic lymphocytes. J
Immunol., 2005 Jul l;175(l):82-9.
Caplen, N.1, Parrish, S., Imani, F., Fire, A. & Morgan, R.A. Specific inhibition of
gene expression by small double stranded RNAs in invertebrate and vertebrate
systems. Proc. Natl. Acad. Sci. USA, 2001, 98: 9742-9747.
Cheewatrakoolpong B, Gilchrest H, Anthes JC, Greenfeder S. Identification and
characterization of splice variants of the human P2X7 ATP channel. Biochem
Biophys Res Commun., 2005 Jun 24;332(1): 17-27.
Chessell IP, Hatcher JP, Bountra C, Michel AD, Hughes JP, Green P, Egerton J,
Murfin M, Richardson J, Peck WL, Grahames CB, Casula MA, Yiangou Y, Birch R,
Anand P, Buell GN. Disruption of the P2X7 purinoceptor gene abolishes chronic
inflammatory and neuropathic pain. Pain, 2005 Apr;114(3):386-96.
Cook, S.P. & McCleskey, E.W. Cell damage excites nociceptors through release
of cytosolic ATP. Pain, 2002, 95: 41-^7.
Cotrina, M.L et al. Connexlns regulate calcium signalling by controlling ATP
release. Proc. Natl, Acad. Sci. USA, 1998,.95:15735-15740.
Deuchars, SA et al. Neuronal P2X7 receptors are targeted to presynaptic
terminals in the central and peripheral nervous systems. J. Neurosci., 2001, 21:
7143-7152.
Di Wgilto, F., Chiozzi, P., Ferrari, D., Falzoni, S., Sanz, 3. M., Morelli, A., Torboli,
M., Bolognesf, G., and Barfeordl, O. R. Nudeotkje receptors: an emerging family
of regulatory molecules in blood cells. Blood, 2001,97, 587-600.
Di Virgilio, F. et al. Cytolytlc P2X purinoceptors. Cell Death Differ., 1998, 5:
191-199.
Du, S. et al. Calcium influx and activation of calpain I mediate acute reactive
gliosis in injured spinal cord. Exp. Neurol., 1999,157: 96-105.
EI-Moatassim, C., and Dubyak, G. R, Dissociation of the pore-forming and
phospholipase D activities stimulated via P2z purinergic receptors in BAC1.2F5
macrophages. Product inhibition of phospholipase D enzyme activity J. Biol.
Chem., 1993, 268,15571-15578.
Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21- and
22-nucleotlde RNAs. Genes Dev., 2001,15(2): 188-200.
Fam, S.R., Gallagher, CJ. & Salter, M.W. P2Y(1) purinoceptor-mediated Ca2+
signalling and Ca2+ wave propagation in dorsal spinal cord astrocytes. 1
Neurosci., 2000, 20: 2800-2808.
Ferrari, D., Los, M., Bauer, M. K. A., Vandenabeele, P., Wesselborg, S., and
Schulze-Osthoff, K. P2Z purinoreceptor iigation induces activation of caspases
with distinct roles in apoptotic and necrotic alterations of cell death. FEBS Lett.,
1999, 447, 71-75.
Ferrari, D., La Sala, A., Chiozzi, P., Morelli, A., Falzoni, S., Girolomoni, G., Idzko,
M., Dichmann, S., Norgauer, J. and DI Virgilio, F. The P2 purinergic receptors of
human dendritic cells: identl®cation and coupling to cytokine release. FASEB 1,
2000, 14:2466.
Reids, R.D. & Stevens-Graham, B. New insights into neuron-glia
communication. Science, 2002,298: 556-562.
Rre A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and
specific genetic interference by double stranded RNA in Caenorhabditis elegans.
Nature, 1998, 391(6669):806-11.
Franke H, Gunther A, Grosche 3, Schmidt R, Rossner S, Reinhardt R, Faber-
Zuschratter H, Schneider D, Illes P. P2X7 receptor expression after ischemia in
the cerebral cortex of rats. 3 Neuropathol Exp Neural., 2004 3ul; 63(7):686-99.
Gargett, C. E., Cornish, E. 3., and Wiley, 3. S. Phospholipase D activation by
P2Z-purlnoceptor agonists in human lymphocytes is dependent on bivalent
cation influx. Biochem. 1,1996, 313, 529-535.
Ge Q, McManus MT, Nguyen T, Shen CH, Sharp PA, Elsen HN, Chen J. RNA
interference of influenza virus production by directly targeting mRNA for
degradation and indirectly inhibiting all viral RNA transcription. Proc Nati Acad
Sci U S A., 2003; 100(5):2718-23.
Gil 3, Esteban M. Induction of apoptosis by the dsRNA-dependent protein kinase
(PKR): mechanism of action. Apoptosis, 2000, 5(2):107-14.
Greig AV, Linge C, Healy V, Lim P, Clayton E, Rustin MH, McGrouther DA,
Burnstock G. Expression of purinergic receptors in non-melanoma skin cancers
and their functional roles in A431 cells. 3 Invest Dermatol., 2003a, 121(2):315-
27.
Greig AV, Linge C, Terenghi G, McGrouther DA, Burnstock G. Purinergic
receptors are part of a functional signaling system for proliferation and
differentiation of human epidermal keratinocytes. j Invest Dermatol., 2003b,
120(6): 1007-15.
Grosshans H, Slack F3. Micro-RNAs: small is plentiful. 3 Cell Biol, 2002,
156(1): 17-21.
Gu, B., Bendall, L 3., and Wiley, 3. S. Adenosine Triphosphate-Induced
Shedding of CD23 and L-Selectin (CD62L) From Lymphocytes Is Mediated by
the Same Receptor but Different Metalloproteases. Blood, 1998, 92, 946-951.
Guthrie et al. ATP released from astrocytes mediates glial calcium waves. 3.
NeuroscL, 1999,19: 520-528.
Hart, D. N. J. Dendritic cells: unique leukocyte populations which control the
primary immune response. Blood, 1997, 90:3245.
Humphreys, B. D., Rice, 3., Kertesy, S. B., and Dubyak, G. R. Stress-activated
Protein Kinase/JNK Activation and Apoptotic InductkNfl by the Macrophage P2X7
Nucleotide Receptor. 3. Bfol. Chem., 2000, 275, 26792-26798.
3amieson/ G. P., Snook, M. B., Thurlow, P. 3., and Wiley, 3. S. Extracellular ATP
causes of loss of L-selectin from human lymphocytes via occupancy of P2Z
purinocepters. 3. Cell. Physiol., 1996,166, 637-642.
Kawamura H, Sugiyama T, Wu DM, Kobayashi M, Yamanishi S, Katsumura K,
Puro DG. ATP: a vasoactive signal in the pericyte-containing microvasculature
of the rat retina. 3 Physiol., 2003 Sep 15;551(Pt 3):787-99. Epub 2003 3ul 22.
Mutini C, Falzoni S, Ferrari D, Chiozzi P, Morelii A, Barlcordi OR, Collo G,
Ricciardl-Castagnoli P, Di Virgilio F. Mouse dendritic cells express the P2X7
purinergic receptor: characterization and possible participation in antigen
presentation. 3 Immunol., 1999 Aug 15;163(4):1958-65.
Neary, 3.T. et al. Activation of extracellular signal-regulated kinase by stretchinduced
injury in astrocytes involves extracellular ATP and P2 purinergic
receptors. 3. Neurosci., 2003, 23: 2348-2356.
North, R, A. Molecular Physiology of P2X Receptors. Physiol. Rev., 2002, 82,
1013-1067.
Paddison P3, Caudy M, Bernstein E, Hannon G3, Conklln DS. Short hairpin
RNAs (shRNAs) induce sequence-specific silencing in mammalian ceils. Genes
Dev., 2002, 16(8):948-58.
Pannicke T, Fischer W, Bledermann B, Schadlich H, Grosche 3, Faude F,
Wiedemann P, Allgaier C, Illes P, Burnstock G, Reichenbach A. P2X7 receptors
in Muller glial cells from the human retina. 3 Neuroscl., 2000 Aug 15;
20(16):5965-72.
Parvathenani LK, Tertyshnikova S, Greco CR, Roberts SB, Robertson B,
Posmantur R. P2X7 mediates superoxlde production in primary microglia and is
up-regulated in a transgenic mouse model of Alzheimer's disease. 3 Biol Chem.,
2003 Apr 11;278(15): 13309-17.
Scemes, E., Suadicani, S.O. & Spray, D.C. Intercellular communication in spinal
cord astrocytes: fine tuning between gap junctions and P2 nucleotide receptors
in calcium wave propagation. 1 Neurosci., 2000, 20: 1435-1445.
Sluyter, R., and Wiley, 3. S. Extracellular adenosine 5'~triphosphate induces a
loss of CD23 from human dendritic cells via activation of P2X7 receptors. Int.
Immunol., 2002,14,1415-1421.
Solini A, Chiozzi P, Morelli A, Adinolfi E, Rlzzo R, Baricordi OR, Di Virgilio F.
Enhanced P2X7 activity in human fibroblasts from diabetic patients: a possible
pathogenetlc mechanism for vascular damage in diabetes. Arterioscler Thromb
Vase Biol., 2004 3ul;24(7): 1240-5.
Stockwin, L H., McGonagle, D., Martin, I. G, and Blair, G, E. Dendritic ceils:
immunological sentinels with a central role in health and disease. Immunol. Cell
Biol., 2000, 78:91.
Taylor AL, Schwlebert LM, Smith 33, King C, 3ones 3R, Sorscher E3, Schwiebert
EM. Epithelial P2X purinergic receptor channel expression and function. 3 Clln
Invest., 1999 Oct;104(7):875-84.
Tuschl T, Zamore PD, Lehmann R, Bartel DP, Sharp PA. Targeted mRNA
degradation by double-stranded RNA in vitro. Genes Dev., 1999; 13(24):3191-
7.
Verhoef PA, Estacion M, Schilling W, Dubyak GR. P2X7 receptor-dependent
blebbing and the activation of Rho-effector kinases, caspases, and IL-1 beta
release. J Immunol., 2003 Jun 1; 170(11):5728-38.
Virginio, C., MacKenzie, A., North, R. A., and Surprenant, A. Kinetics of cell
lysis, dye uptake and permeability changes in cells expressing the rat P2X7
receptor. J. Physiol. (Lond.), 1999, 519, 335-346.
Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, Li P, Xu Q, Liu QS,
Goldman SA, Nedergaard M. P2X7 receptor inhibition improves recovery after
spinal cord injury. Nat Med., 2004 Aug;10(8):821-7. Epub 2004 Jul 18.
Wlanny F, Zernicka-Goetz M. Specific interference with gene function by
double-stranded RNA in early mouse development Nat Cell Biol., 2000,
2(2):70-5.
Williams BR. Role of the double-stranded RNA-activated protein kinase (PKR) in
cell regulation. Btochem Soc Trans, 1997r 25(2):509-13.
Zhang X, Zhang M, Laties AM, Mitchell CH. Stimulation of P2X7 receptors
elevates Ca2+ and kills retinal ganglion cells. Invest Ophthalmol Vis Sci., 2005
Jun; 46(6):2183-91.
Zhang XJ, Zheng GG, Ma XT, Yang YH, Li G, Rao Q, Nie K, Wu KF. Expression
of P2X7 in human hematopoietic cell lines and leukemia patients. Leuk Res.,
2004 Dec;28(12):l313-22.




CLAIMS
1. Use of siNA in the preparation of a medicament for use in a method of
treatment of a condition characterised by increased expression and/or activity
of P2RX7, the method comprising downregulating expression of P2RX7 in a
patient.
2. The use of claim 1 wherein the condition is selected from the group
comprising neuronal degeneration, reperfusion or ischemia in stroke or heart
attack, Alzheimer's disease, inflammatory diseases (such as rheumatoid
arthritis, osteoarthritis, asthma, rhinitis, chronic obstructive pulmonary disease
(COPD), Inflammatory bowel disease (IBD) such as Crohn's disease), allergies,
autoimmune diseases, cancer (such as leukaemia or non-melanoma skin
cancer), skin-related conditions (such as psoriasis, eczema, alopecia), retinal
diseases and treatment of pain of neuropathic and inflammatory origin.
3. The use of any preceding daim wherein the siNA is siRNA.
4. The use of claim 3 wherein the siRNA is dsRNA.
5. The use of claim 3 wherein the siRNA is shRNA.
6. The use of any preceding claim wherein the siNA comprises a modified
oligonucleotide.
7. The use of any preceding claim wherein the siNA is administered topically to
a patient.
8. The use of any preceding claim wherein a plurality of species of siNA are
used.
9. The use of claim 8 wherein said plurality of species are targeted to the same
mRNA species.
10. The use of daim 8 wherein said plurality of species are targeted to different
mRNA species.
11. The use of any preceding claim wherein the siNA is targeted to a sequence
selected from SEQ ID 1 to SEQ ID 109.
12. The use of any preceding daim wherein the siNA Is targeted to a splice
form of P2RX7 selected from the group having GenBank Accession Numbers
NMJ302562, NM_177427, AY847298, AY847299, AY847300, AY847301,
AY847302, AY847303, AY847304.
13. A method of treatment of a disease condition characterised by increased
expression and/or activity of P2RX7, comprising administering siNA to
downregulate expression of P2RX7 gene In a patient
14. The method of claim 13, wherein the disease condition is selected from the
group comprising neuronal degeneration, reperfuslon or Ischemia in stroke or
heart attack, Alzheimer's disease, inflammatory diseases (such as rheumatoid
arthritis, osteoarthritis, asthma, rhinitis, chronic obstructive pulmonary disease
(COPD), inflammatory bowel disease (IBD) such as Crohn's disease), allergies,
autoimmune diseases, cancer (such as leukaemia or non-melanoma skin
cancer), skin-related conditions (such as psoriasis, eczema, alopecia), retinal
diseases and treatment of pain of neuropathic and inflammatory origin.
15. The method of claim 13, wherein the disease condition is neuronal
degeneration.
16. The method of daim 13, wherein the disease condition is reperfusion or
ischemia in stroke or heart attack.
17. The method of daim 13, wherein the disease condition is Alzheimer's
disease.
18. The method of daim 13, wherein the disease condition is an inflammatory
disease (such as rheumatoid arthritis, osteoarthritis, asthma, rhinitis, chronic
obstructive pulmonary disease (COPD), inflammatory bowel disease (IBD) such
as Crohn's disease).
19. The method of daim 13, wherein the disease condition is an allergy.
20. The method of daim 13, wherein the disease condition is an autoimmune
disease.
21. The method of daim 13, wherein the disease condition is cancer (such as
leukaemia or non-melanoma skin cancer).
22. The method of claim 13, wherein the disease condition is a skin-related
condition (such as psoriasis, eczema, alopecia).
23. The method of claim 13, wherein the disease condition is a retinal disease.
24. The method of claim 13, wherein the disease condition is pain of
neuropathic and inflammatory origin.
25. The method of claim 13 wherein the siNA is siRNA.
26. The method of claim 25 wherein the siNA is dsRNA.
27. The method of claim 25 wherein the siNA is shRNA.
28. The method of claim 13 wherein the siNA comprises a modified
oligonucleotide.
29. An isolated siNA molecule for use in the treatment of a disease condition
characterized by Increased expression and/or activity of P2RX7, the siNA being
complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID
109.
30. Use of an isolated siNA molecule having a sequence which is
complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID 109
in the preparation of a medicament for the treatment of a disease condition.
31. A pharmaceutical composition comprising siNA having a sequence which is
complementary to a nucleotide sequence selected from SEQ ID 1 to SEQ ID
109.
32. Method for the treatment of a condition associated with, or mediated by, an
increase of extra cellular calcium, comprising downregulation of P2RX7 gene
expression by administering to a subject a pharmaceutically effective
preparation for causing RNAi.
33. The method of claim 32, in which the condition is associated with reduced
blood flow to the brain and other CNS tissue, or associated with instances of a
temporary break in blood supply to the brain or to other CNS tissue.
34. The method of claim 32 or 33, in which the condition is an ischaemic
disease, an anoxic episode, an injury to the brain and other parts of the CNS
caused by trauma or other injury, a blow to the head, or a spinal injury, a
thromboembolic or haemorrhagic stroke, a cerebral vasospasm, hypogiycaemia,
cardiac arrest, status epitepticus, perinatal asphyxia, anoxia, cerebral trauma,
lathyrism, Alzheimer's disease, Parkinson's Disease and Huntmgton's disease,
cerebral ischaemia or cerebral infarction, ischaemlc, hypoxic or anoxic brain
damage, spinal cord injury, tissue ischaemia and reperfusion injury in a
mammal at risk for such damage.
35. A method for the detection of a pharmacological activity in a biological
sample comprising administering a sample to a cell, and determining a change
in activity of P2RX7.
36. The method of daim 35, wherein a change In activity is determined by
comparing a cell In which P2RX7 gene expression has been downregulated with
a normal cell to which the sample has been administered.
37. The method of daim 36 In which P2RX7 downregulation is effected using
siNA.
38. The method of any of daims 35 to 37, wherein the biological sample Is of
marine origin.
39. An isolated siNA molecule, a pharmaceutical composition, a method of treatment
and use of siNA molecule in down regulating the expression of P2RX7 receptors thereof
substantially as herein described with reference to the foregoing examples and
accompanying figures.

Documents:

2298-delnp-2007-Abstract-(07-03-2013).pdf

2298-delnp-2007-abstract.pdf

2298-delnp-2007-Claims-(07-03-2013).pdf

2298-delnp-2007-claims.pdf

2298-delnp-2007-Correspondence Others-(07-03-2013).pdf

2298-delnp-2007-Correspondence Others-(07-08-2012).pdf

2298-delnp-2007-correspondence-others-1.pdf

2298-delnp-2007-correspondence-others.pdf

2298-delnp-2007-description (complete).pdf

2298-delnp-2007-Description (Completer)-(07-03-2013).pdf

2298-delnp-2007-drawings.pdf

2298-delnp-2007-Form-1-(07-08-2012).pdf

2298-delnp-2007-form-1.pdf

2298-delnp-2007-form-18.pdf

2298-delnp-2007-Form-2-(07-03-2013).pdf

2298-delnp-2007-form-2.pdf

2298-delnp-2007-Form-3-(07-03-2013).pdf

2298-delnp-2007-form-3.pdf

2298-delnp-2007-form-5.pdf

2298-delnp-2007-GPA-(07-08-2012).pdf

2298-delnp-2007-pct-101.pdf

2298-delnp-2007-pct-210.pdf

2298-delnp-2007-pct-304.pdf

2298-delnp-2007-Petition-137-(07-03-2013).pdf


Patent Number 260264
Indian Patent Application Number 2298/DELNP/2007
PG Journal Number 16/2014
Publication Date 18-Apr-2014
Grant Date 16-Apr-2014
Date of Filing 23-Mar-2007
Name of Patentee SYLENTIS S.A.U.
Applicant Address Calle Jose Abascal 2 E-28003 MADRID, SPAIN.
Inventors:
# Inventor's Name Inventor's Address
1 ROMAN, JOSE P. C/ ARGENTINA, 30. 6ºC, E-01009 VITORIA SPAIN.
2 GASCON, IRENE AVDA/ RAMON Y CAJAL, 1. 5º LZDA, E-28016 MADRID, SPAIN.
3 GONZALEZ DE BUITRAGO, GONZALO C/ LEON GIL DE PALACIO, 5. ESC. DCHA, 6ºA., E-28007 MADRID, SPAIN.
4 JIMENEZ, ANA I. C/ CAZADORES, 15, ALCORCON, E-28922 MADRID SPAIN.
5 SESTO, ANGELA C/ ANDRES MELLADO, 85. ESC. LZDA 6ºB, E-28003 MADRID, SPAIN.
PCT International Classification Number C12N 15/11
PCT International Application Number PCT/GB2005/050139
PCT International Filing date 2005-08-30
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 0419295.1 2004-08-31 U.K.
2 0504057.1 2005-02-28 U.K.