throbber
Citation: Molecular Therapy–Nucleic Acids (2012) 2, e8; doi:10.1038/mtna.2011.9
`© 2012 American Society of Gene & Cell Therapy All rights reserved 2158-3188/11
`www.nature.com/mtna
`
`COMMENTARY
`The Business of RNAi Therapeutics in 2012
`
`Dirk Haussecker1
`
`Molecular Therapy–Nucleic Acids (2012) 2, e8; doi:10.1038/mtna.2011.9; published online 7 February 2012
`
`INTRODUCTION
`
`In its decade in existence, commercial RNA interference
`(RNAi) therapeutics development has seen great financial
`volatility. The causes of this volatility are broadly shared with
`what has been observed on other technology frontiers such
`as gene therapy in the case of drug development, with the
`amplitude of the volatility magnified or moderated by macro-
`economic factors. Volatility poses challenges especially for
`financially exposed small biotechnology companies, the core
`translational force of the industry, to establish the platform
`and develop drugs in a process that takes at least 15 years
`to bear fruits in the form of approved drugs and depends on
`the complex interactions between a diverse set of investors.
`Even small disruptions can have big repercussions leading
`to both euphoria and capitulation which can be equally dam-
`aging to the long-term health of a sector.
`This commentary is directed at companies already involved
`in RNAi therapeutics development or those interested in
`entering the space. By analyzing the forces that shape the
`business of RNAi therapeutics at the start of 2012 it aims
`to uncover key opportunities for value creation. It may also
`help investors identify related investment opportunities and
`inventors commercialize their intellectual property (IP). For a
`review of the fundamental business case for RNAi therapeu-
`tics, the reader is referred to an earlier article on the topic.1
`
`RNAi THERAPEUTICS BUSINESS TRENDS IN
`HISTORICAL PERSPECTIVE
`
`The business of RNAi therapeutics has just entered its fourth
`phase. The first, discovery phase (2002–05) was defined by
`the early adopters of RNAi as a therapeutic modality follow-
`ing the discovery of RNAi in human cells.2 These were small,
`risk-taking biotechnology companies such as Ribozyme
`Pharmaceuticals (aka Sirna Therapeutics), Atugen (aka
`Silence Therapeutics) and Protiva (aka Tekmira). As much
`as they may have believed in the potential of RNAi thera-
`peutics, their strategic reorientation was also a gamble on a
`technology with considerable technical uncertainties in order
`to turn around declining business fortunes by leveraging their
`nucleic acid therapeutics know-how to become leaders in a
`potentially disruptive technology. For example, exploration of
`in vivo gene knockdown had only just begun, not to speak
`
`of knockdown in larger animals following systemic delivery.
`This phase also saw the founding of Alnylam Pharmaceutical
`based on the idea of cornering the IP on the molecules that
`mediate RNAi (RNAi triggers) so that it may finance its own
`drug development by collecting a toll from all those engaged
`in RNAi therapeutics.
`(“Big
`Until
`then,
`larger pharmaceutical companies
`Pharma”) saw the value of RNAi largely as a research tool
`only. This, however, changed quickly when a few of them,
`including Medtronic, Novartis, and Merck, were seen by their
`peers to take an interest in RNAi as a therapeutic modality.
`The situation seemed reminiscent of monoclonal antibodies
`which had just established themselves as the major value
`creator in the pharmaceutical industry, but where Big Pharma
`was thought to be paying the price for having watched from
`the sidelines for too long. Another factor for Big Pharma’s
`surging RNAi therapeutics interest, the defining feature of
`the second, boom phase of RNAi therapeutics (2005–08),
`was the impending patent cliff and the hope that the technol-
`ogy would mature in time to soften its financial impact.
`A bidding war, largely for access to potentially gate-
`keeping RNAi trigger IP erupted. Most notably, Merck and
`Roche paid US$1.1B for acquiring Sirna Therapeutics and
`US$300M+ for a limited platform license from Alnylam,
`respectively. These deals were only rivaled in attention by
`the award of a Nobel Prize to Andrew Fire and Craig Mello
`for their seminal discovery of double-stranded RNA (dsRNA)
`as the trigger of RNAi. The industry naturally did not mind
`the attention and in some cases fanned the fire by raising
`unrealistic expectations. This atmosphere also gave rise to
`controversial publications in high-profile journals which lent
`credence to the mistaken notion that the technical barriers to
`exploiting the RNAi trigger IP would be low.3,4 Consequently,
`most Big Pharma companies had a stake in the technology.
`Yet, the US$2.5B–3.5B in investments largely failed to formu-
`late sound strategies for the real technical challenges such
`as delivery. Symptomatic for the times, the financial markets
`similarly failed to realize the value of truly enabling technolo-
`gies: in the 2 weeks following the publication of a seminal
`paper on systemic small interfering RNA (siRNA) delivery
`by Protiva (now Tekmira) and Alnylam on 26 March 2006,5
`Alnylam’s share price would decline by over 10%.
`It is therefore perhaps not surprising that this period of high
`expectations and blockbuster deals was followed by general
`
`1Department of Medical Biotechnology, College of Natural Sciences, Dongguk University, Seoul, Korea
`Correspondence: Dirk Haussecker, Department of Medical Biotechnology, College of Natural Sciences, Dongguk University, 26, Pil-dong 3, Jung-gu, Seoul 100-715,
`Korea.
`E-mail: dirk.haussecker@gmail.com
`
`PROTIVA - EXHIBIT 2015
`Moderna Therapeutics, Inc. v. Protiva Biotherapeautics, Inc.
`IPR2018-00739
`
`

`

`2
`
`
`RNAi Therapeutics Business 2012
`
`backlash (2008–2011), the financial consequences of which
`were exacerbated by global economic turmoil and health-
`care rationing in the West. Big Pharma quickly realized their
`mistake of putting IP before enablement as they scrambled
`to scout for delivery technologies and found the majority of
`them not to live up to their claims.6 Roche, a year after their
`IP license from Alnylam, felt compelled to pay US$125M for
`Dynamic PolyConjugates from Mirus Bio, one of the more
`promising and differentiated delivery technologies, for which,
`however, significant risks related to translation into organ-
`isms beyond rodents and manufacturing/scale-up remained.
`Contributing to buyer’s remorse was the ageing and rapidly
`eroding gate-keeping potential of the RNAi trigger IP that
`had been the focus of their original investments.
`As much as delivery, it was the potential of certain RNAi for-
`mulations to stimulate innate immunity that caused much of
`the scientific angst that contributed to the deteriorating busi-
`ness sentiment in 2008.7,8 It almost came to be assumed that
`an in vivo RNAi efficacy claim was in fact an innate immuno-
`stimulatory artefact. Importantly, this suspicion extended to
`the preclinical data that formed the rationale for the industry’s
`lead clinical candidates in wet age-related macular edema
`(Acuity/Opko’s Cand5, Merck/Allergan’s Sirna-027/AGN-
`745, Quark/Pfizer’s PF-4523655)9,10 and respiratory viral
`infection (Alnylam’s ALN-RSV01),11 approaches which inci-
`dentally did not involve specific delivery chemistries. Mak-
`ing matters worse still, innate immune stimulation is a safety
`issue. Although today innate immunostimulatory potential is
`widely considered to be manageable through chemical modi-
`fication and choice of RNAi trigger structure, the reputational
`damage persists.
`Suffering from RNAi-specific scientific and credibility issues
`and with first drug approvals still years away, RNAi therapeu-
`tics was among the first to feel the cost-cutting axe at com-
`panies like Pfizer, Merck, Abbott Labs, and Roche which all
`started to suffer from patent expirations, drug approval and
`productivity issues, worsening drug reimbursement climates,
`and the general loss of confidence in their innovative abili-
`ties. Particularly the exit of Roche from in-house RNAi thera-
`peutics development sent shockwaves through the industry.
`Having invested heavily in the technology only 2–3 years ago
`and being considered an innovation bellwether within Big
`Pharma, Roche’s decision in late 2010 found a number of
`imitators among Big Pharma and has been functioning as a
`major barrier to new investments in RNAi therapeutics.
`The backlash, however, also had cleansing effects which
`form the basis for the 4th, recovery phase of RNAi therapeu-
`tics (2011–present). As a result of the financial restrictions
`and increased scientific scrutiny, there has been an overall
`increase in the quality of the science. RNAi therapeutics
`has also become less of a target for the quick-rich biotech
`schemes that constantly chase the next hot area in drug
`development. This quality shift is most evident in the evo-
`lution of the RNAi therapeutics clinical pipeline which has
`become more and more populated with candidates based
`on sound scientific rationales, especially in terms of delivery
`approaches and anti-immunostimulatory strategies. For the
`recovery, however, to firmly take root and for the long-term
`health of the industry, it is important for the current clinical
`dataflow to bring back investors.
`
`RNAi THERAPEUTICS ASSETS
`
`One measure for the health of an industry is in accounting
`its assets. These are also at the center of business activity.
`Because drugs are the ultimate objective of RNAi therapeu-
`tics and because of the significant de-risking that occurs dur-
`ing drug development, the clinical and late-stage preclinical
`pipeline weighs heavy. Equally important at this relatively
`early stage are the technologies that enable candidate devel-
`opment and drive platform efficiencies. These technologies
`need to be protected by patents or trade secrets for individ-
`ual companies to capture their full value.
`
`RNAi therapeutics development pipeline. As of the 2008
`review,1 there were eight candidates in clinical development
`(Table 1). What is noticeable is that most of them were local
`RNAi approaches that today would most likely not enter devel-
`opment due to uncertain scientific rationale or safety: naked
`delivery, in some cases with unmodified synthetic RNAi trig-
`gers (Cand5, Sirna-027, RTP-801i, ALN-RSV01, TD-101),
`liposomal delivery of a DNA-directed RNAi (ddRNAi) can-
`didate which could have been predicted to be inadequate
`for antiviral applications and was all but assured to cause
`immune stimulation (NucB1000),12 or first-generation ddRNAi
`expression systems subsequently13 found to frequently cause
`cellular toxicity (rHIV-shI-TAR-CCR5RZ; possibly NucB1000).
`Not surprisingly, many of these programs were either termi-
`nated, or their future development is doubtful. Among the
`latter, there is hope that Quark/Pfizer’s PF-4523655 and
`Alnylam’s ALN-RSV01 can still make it to market as long as
`they show appropriate safety and efficacy even though their
`value to RNAi therapeutics would be limited given the wide-
`spread skepticism about their mechanism of action.
`Since 2008, the development pipeline has not only grown
`in size (18 active clinical candidates today), but more impor-
`tantly it has improved in quality concomitant with a shift from
`local to systemic delivery: 7 of the 14 new clinical candidates
`since 2008 were delivered systemically, compared to only 1
`of the 8 before. This is largely the result of the clinical entry
`of the most advanced systemic delivery platforms, stable
`nucleic acid lipid particles (SNALP) and AtuPLEX. SNALP
`alone accounts for six clinical candidates (ALN-VSP02, TKM-
`ApoB, ALN-TTR01, TKM-PLK1, ALN-PCS02, TKM-EBOLA)
`and one more is expected to enter the clinic in the near future
`(ALN-TTR02).
`Given that the value of a given drug candidate is dynamic
`and can dramatically change with each new data point—such
`as a clinical trial result or even change in regulatory policy—
`it is beyond the scope of this commentary to determine the
`market value of the RNAi development pipeline. Some candi-
`dates, however, have been licensed which makes their market
`value easier to assess. Quark Pharmaceuticals for example
`has been quite successful in licensing its compounds. As of 31
`December 2010, Pfizer had invested $52.5M in PF-4523655
`which is in late phase II development for wet age-related mac-
`ular edema and diabetic macular edema. Quark moreover is
`eligible to receive substantial future milestones and royalties.14
`Still, the value of PF-4523655 has become highly uncertain
`after phase II study results suggested that PF-4523655 faces
`an uphill battle before it can be a commercially viable drug.
`
`Molecular Therapy–Nucleic Acids
`
`

`

`Table 1 RNAi therapeutics clinical pipeline
`
`Year of
`IND/CTA
`
`
`Candidate
`
`
`Indication
`
`2004
`2004
`2005
`
`2007
`
`2007
`2007
`
`2007
`
`2008
`2008
`
`2008
`
`2008
`
`2008
`
`2009
`
`2009
`
`2009
`2009
`
`2009
`
`2009
`
`2010
`
`2010
`
`2011
`
`2011
`
`2011
`
`Wet AMD, diabetic macular edema
`Cand5
`Sirna-027/AGN-745 Wet AMD
`ALN-RSV01
`RSV infection
`
`DGFi
`
`PF-4523655
`rHIV-shl-TAR-
`CCR5RZ
`NucB1000
`
`Acute kidney injury, delayed
`graft function
`Wet AMD, diabetic macular edema
`HIV infection
`
`Hepatitis B viral infection
`
`HBV RNAs
`
`TD101
`Therapeutic vaccine
`
`Pachyonychia congenita
`Metastatic melanoma
`
`Mutant keratin
`Immunoproteasome
`
`Excellair
`
`Asthma
`
`Syk kinase
`
`CALAA-01
`
`ALN-VSP02
`
`Atu027
`
`QPI-1007
`
`SYL040012
`TKM-ApoB
`
`bi-shRNAfurin/
`GMCSF
`ALN-TTR01
`
`siG12D LODER
`
`TKM-PLK1
`
`CEQ508
`
`ALN-PCS02
`
`Nonresectable or metastatic solid
`tumors
`Liver cancer, cancer with liver
`involvement
`Advanced solid tumors
`
`Chronic nerve atrophy, nonarteritic
`ischemic optic neuropathy
`Intraocular pressure and glaucoma
`Hypercholesterolemia
`
`M2 subunit of ribonucleotide
`reductase
`VEGF, KSP
`
`PKN3
`
`Caspase 2
`
`(cid:66)-Adrenergic receptor 2
`Apolipoprotein B
`
`Ovarian cancer, advanced
`melanoma
`Transthyretin amyloidosis
`
`Operable pancreatic ductal
`adenocarcinoma
`Solid cancers and lymphoma
`
`Furin
`
`Transthyretin
`
`Mutated KRAS
`
`Polo-like kinase 1
`
`Familial adenomatous polyposis/
`colon cancer prevention
`Hypercholesterolemia
`
`-Catenin
`
`PCSK9
`
`TKM-EBOLA
`
`Ebola infection (biodefense)
`
`Viral RNA
`
`
`RNAi Therapeutics Business 2012
`
`3
`
`
`Target
`
`VEGF
`VEGF-R1
`Viral RNA
`
`p53
`
`RTP801/REDD1
`Viral RNA and host factors
`
`
`Delivery
`
`Intravitreal needle injection (retina; local)
`Intravitreal needle injection (retina; local)
`Inhalation of unformulated siRNAs
`(lung epithelium; local)
`Intravenous naked siRNA (proximal
`tubule cells; systemic)
`Intravitreal needle injection (retina; local)
`Lentiviral (hematopoietic stem cells;
`ex vivo)
`Liposomal plasmid (hepatocytes;
`systemic)
`Intradermal needle injection (skin; local)
`Electroporation (autologous monocytes;
`ex vivo)
`Inhalation of unformulated siRNAs
`(lung epithelium; local)
`RONDEL (solid tumor cells; systemic)
`
`SNALP liposome (hepatocytes;
`systemic)
`AtuPLEX lipoplex (vascular endothelial
`cells; systemic)
`Intravitreal needle injection
`
`Eye drop (ciliary epithelial cells; local)
`SNALP liposome (hepatocytes;
`systemic)
`Electroporation plasmid (autologous
`tumor samples; ex vivo)
`SNALP liposome (hepatocytes;
`systemic)
`LODER local drug elution
`
`SNALP liposomal (solid tumor cells;
`systemic)
`Bacterial (mucosal layer of small and
`large intestine; oral)
`SNALP liposome (hepatocytes;
`systemic)
`SNALP liposome (hepatocytes and
`phagocytes; systemic)
`
`Intradermal needle injection (skin; local)
`Lentiviral transduction transduction
`(hematopoietic stem cells; ex vivo)
`
`Select preclinical candidates
`
` 2012 (est.)
` 2012 (est.)
`
`RXI-109
`To be named
`
`Dermal scarring
`HIV infection
`
`CTGF
`CCR5
`
`Abbreviations: AMD, age-related macular edema; CTGF, connective tissue growth factor; GMCSF, granulocyte-macrophage colony-stimulating factor; HBV,
`hepatitis B virus; KSP, kinesin spindle protein; PKN3, protein kinase N3; RSV, respiratory syncytial virus; shRNA, small hairpin RNA; siRNA, small interfering
`RNA; SNALP, stable nucleic acid lipid particles; VEGF, vascular endothelial growth factor.
`
`Quark also sold an option for an exclusive license to its sec-
`ond-most advanced candidate, QPI-1002, then in phase I for
`acute kidney injury and delayed graft function, for a remark-
`able US$10M fee to Novartis. The market value of the only
`other partnered candidate in clinical development, ALN-
`RSV01, has decreased considerably as its target population
`has shrunk drastically and after Alnylam’s partners for this
`drug candidate, Kyowa Hakko and Cubist Pharmaceuticals,
`have distanced themselves from it despite having invested
`more than US$35M in upfront alone.
`The remaining value of the clinical pipeline largely rests on
`three oncology candidates (ALN-VSP, Atu027, TKM-PLK1)
`
`and the SNALP-enabled ALN-PCS02 for hypercholester-
`olemia and ALN-TTR01/02 for transthyretin amyloidosis.
`This judgment is based on delivery that has been de-risked
`to some extent for these candidates, almost nonexistent tar-
`get risks for three of them (TTR, PLK1, PCS02), and the fact
`that they all represent highly differentiated approaches for
`diseases of considerable unmet medical needs. Moreover,
`there exist early biomarker opportunities for two of them
`(TTR, PCS). Should these biomarker read-outs demonstrate
`effective target gene knockdown in their phase I studies, their
`value would increase considerably, possibly pegging their
`upfront partnering value in the high double-digit millions with
`
`Molecular Therapy–Nucleic Acids
`
`

`

`4
`
`
`RNAi Therapeutics Business 2012
`
`the potential to generate substantially more revenues down-
`stream. In the case of ALN-VSP and Atu027, early clinical
`data are already supportive of further development with the
`sponsors hoping to license these compounds in 2012.
`Although having only just entered clinical development,
`TKM-EBOLA may actually be the pipeline asset with the
`highest net present value in the industry. This is because
`the full development of this biodefense candidate is being
`funded under a US$140M contract from the US Department
`of Defense. This contract allows Tekmira to not only earn
`incentive fees and profit from eventual stockpiling contracts,
`but also to develop the candidate in a way that broadly ben-
`efits the platform on which it was built. Among the other pre-
`clinical pipeline candidates, RXi Pharmaceutical’s RXI-109
`for dermal scarring, and Calimmune’s ddRNAi candidate
`for HIV deserve special mention based on their promising
`preclinical results,15 differentiation, and potential to blaze the
`trail for their respective self-delivering rxRNA and lentiviral
`ddRNAi platforms.
`
`Enabling technologies. As indicated by the evolution of
`the RNAi therapeutics product pipeline, it is the underlying
`technologies, foremost delivery, that are the major value driv-
`ers. Other technologies, however, also add value by reducing
`adverse event risk, and in the case of RNAi trigger innova-
`tion by opening up new therapeutic frontiers.
`Delivery: one cell/tissue type, many indications. The pres-
`ent expansion of the SNALP-based pipeline reflects a fun-
`damental principle of RNAi therapeutics: once a delivery
`technology is found suitable for knocking down genes in a
`given cell/tissue type, any gene can be targeted in that cell/
`tissue type with the possible applications only limited by
`our exploding understanding of disease genetics (Table 2).
`SNALP, Tekmira’s PEG-stabilized monolamellar liposomes
`that encapsulate the RNAi trigger payload in its aqueous
`interior and which are neutrally charged at physiologic pH,
`is furthest developed for knocking down genes expressed in
`the liver, particularly hepatocytes.5 Solid tumor cells,16 sites of
`tissue inflammation, and phagocytic cells,17 however, are also
`suitable targets for SNALP due to their relative accessibility
`and/or natural propensity to take up nanosized particles.
`With the caveat that there is sequence-dependent variabil-
`ity, results from the SNALP-based trials with TKM-ApoB and
`ALN-VSP02 suggest that the SNALP formulations that were
`developed initially have potential for a few indications with
`
`Table 2 Tissues/cell types amenable to therapeutic RNAi today
`
`Tissue/Cell type
`
`Liver (hepatocytes, but also other cell types)
`Vascular endothelial cells
`Solid tumor cells
`Phagocytic cells, including antigen presenting cells
`Skin
`
`Hematopoietic stem cells
`CNS, eye
`
`Delivery
`
`SNALP
`AtuPLEX
`SNALP
`SNALP
`Self-delivering
`rxRNAs
`Lentivirus
`AAV, lentivirus
`
`Abbreviations: AAV, adeno-associated virus; CNS, central nervous
`system; RNAi, RNA interference; SNALP, stable nucleic acid lipid
`particles.
`
`less stringent tolerability and cost requirements. Improve-
`ments in the efficacy and tolerability of SNALP over the last
`5 years,18 however, have significantly widened applicability
`through an expected 100- to 1,000-fold improvement in the
`therapeutic index, and further enhanced the competitive pro-
`file of SNALP by reducing cost and treatment frequencies.
`Symbolizing the value shift from RNAi triggers to delivery,
`Alnylam, which once relied on its RNAi trigger IP for its indus-
`try-leading position, has been sued by Tekmira for scheming
`to unlawfully gain control and ownership over SNALP tech-
`nology and otherwise causing damage to Tekmira’s com-
`petitive position. Somewhat benefitting from this gridlock in
`SNALP is the industry’s second-most advanced systemic
`delivery technology, AtuPLEX by Silence Therapeutics. This
`multilamellar, positively charged, lipid-based formulation
`has proven useful for knocking down genes in the vascular
`endothelium in small and large animal models.19 The phar-
`macokinetic and safety data that emerges from the ongoing
`Atu027 trial (e.g., ASCO 2011 poster presentation) indicate
`this also likely to be the case in humans. With applications
`particularly in oncology (antiangiogenesis) and acute inflam-
`matory conditions (the vascular endothelium as a barrier to
`inflammatory cell infiltration), this technology has garnered
`increased partnership interest. Positively charged lipoplexes,
`in this case delivered by intravesical instillation, may also be
`useful for knocking down genes in the superficial layers of
`the bladder, including malignancies, as suggested by pre-
`clinical data from Marina Biotech.20
`Besides these and other lipid-based delivery technologies,
`there are a number of polymer and conjugate delivery tech-
`nologies in earlier development. What started with largely
`negatively charged RNAi triggers complexed to positively
`charged polymers, an approach frequently associated with
`toxicities,21 polymers appear to be more promising as neutrally
`charged polyconjugates.22 Especially the smaller conjugates
`may be suited for gene knockdown in tissues not accessible
`to the larger lipid-based formulations. Manufacturing chal-
`lenges and biodegradability issues, however, could be caus-
`ing delays in their clinical translation. This appears to be the
`case for the Dynamic PolyConjugates for which Roche paid
`US$125M in 2008, but which Arrowhead Research recently
`acquired for single-digit million US dollars.23
`Smaller than polyconjugates, simple conjugates such
`as the GalNAc-siRNAs (target organ: liver) developed by
`Alnylam may similarly reach a wider range of target cells/
`tissues and could also be amenable to subcutaneous admin-
`istration. Potency improvements, however, are required to
`render them competitive with the more complex formula-
`tions for systemic applications when the target cell/tissue is
`shared. It is in local/localized applications that similar small
`conjugates currently have most utility. A first such program is
`about to enter clinical development with RXi Pharmaceuti-
`cal’s intradermally injected self-delivering rx-RNA RXI-109 for
`dermal scarring. Ocular, central nervous system (intraparen-
`chymal, intrathecal) and respiratory (epithelial) applications
`may similarly benefit from simple conjugate solutions.
`The RNAi trigger versus delivery debate is more balanced
`in ddRNAi technology. This is because delivery technologies
`can be directly borrowed from the field of gene therapy, with
`particularly adeno-associated virus and lentiviral delivery
`
`Molecular Therapy–Nucleic Acids
`
`

`

`
`RNAi Therapeutics Business 2012
`
`5
`
`well suited for a number of central nervous system,24 ocular,25
`and hematopoietic stem cell-related applications.15 Con-
`versely, because ddRNAi is intended for gene silencing over
`extended periods of time following a single administration,
`and adverse reactions due to ddRNAi trigger activity cannot
`easily be reversed, ddRNAi trigger safety is paramount.13
`Some of the delivery technologies above can also be used
`for ex vivo delivery. Here, the delivery challenge is essentially
`reduced to a tissue culture problem by RNAi treating the tar-
`get cells outside the body using transfection, electroporation,
`or viral transduction, before (re-)introducing them into the
`patient. This approach holds particular promise for stem cell-
`based therapeutics15 and therapeutic cancer vaccines.26
`In summary, albeit delivery technologies of clinical and
`commercial maturity are still relatively few in number, today’s
`delivery capabilities already allow for a number of high-quality
`RNAi therapeutics opportunities. This is because each deliv-
`ery technology, once found to be suitable for gene knock-
`down in a given cell/tissue type, can be rapidly expanded
`to many target genes and applications. Control over and
`access to these technologies is critical for RNAi therapeutics
`platform success.
`RNAi triggers: potency matters, but value also in safety
`and new functionalities. One of the main developments in
`the RNAi trigger field has been the realization that many
`RNAs with dsRNA elements can induce RNAi gene silenc-
`ing at least to some degree.27 Together with the weaken-
`ing of Alnylam’s RNAi trigger IP estate in the course of
`the Kreutzer–Limmer (KL) and Tuschl patent prosecutions,
`choice and access to RNAi triggers has become less rate-
`limiting than it was once thought of. It also means that work-
`ing around somebody else’s IP estate alone does not easily
`compensate for deficiencies in scientific performance, espe-
`cially knockdown potency which normally determines both
`the maximal degree and duration of the knockdown. Conse-
`quently, non-Tuschl RNAi triggers should be at least equal
`in potency, if not superior, or offer additional advantages in
`safety and functionality.
`In terms of potency, a single asymmetric instead of sym-
`metrical 3(cid:97) overhangs on the guide strand has been found
`to improve on the knockdown efficacy of Tuschl siRNAs.28
`Potency can also be improved by applying thermodynamic
`design rules such as the Zamore rules to which Silence
`Therapeutics has an exclusive license.29 Although the Dicer-
`substrate RNAi triggers had once been proposed not only to
`fall outside of Alnylam’s RNAi trigger patent estate, but also
`to be more potent than Tuschl siRNAs,30 they may actually be
`a more appropriate example for the value of functional differ-
`entiation by facilitating certain delivery strategies31 and poten-
`tially also by extending the duration of gene silencing.32
`Synthetic small hairpin RNAs can either function as Dicer-
`substrate RNAs or also be smaller in size, yet still trigger RNAi
`(e.g., SomaGenics).32 These single-molecule RNAs have the
`benefit of increased thermodynamic stability which may be
`exploited for the manufacture of RNAi triggers with increased
`dsRNA yield than conventional two-stranded siRNAs33 as
`well as delivery approaches which require single-stranded
`phases during the delivery journey. Shorter small hairpin
`RNAs should also be less prone to induce innate immunity
`and interfere with endogenous small RNA processing. The
`
`latter attributes also apply to the first-generation asymmet-
`ric siRNAs (asiRNA) by Biomolecular Therapeutics which
`are characterized by shorter double-stranded elements
`than those in conventional siRNAs.34 RXi Pharmaceutical’s
`sd-rxRNAs have even shorter double-stranded elements, a
`feature the company claims to be critical for crossing hydro-
`phobic lipid bilayers during delivery. Nevertheless, because
`the success rate of finding potent RNAi triggers may drop
`noticeably for RNAi triggers with such short dsRNA ele-
`ments, these structures should be preferentially contem-
`plated in applications where they can add unique delivery or
`safety benefits.
`The structural flexibility of RNAi triggers has also been
`exploited for increased functionality by having them target
`more than one gene (“multitargeting”). This is particularly
`valuable for treating complex diseases or where resistance
`is an issue (cancer, viral infections). Multitargeting is already
`being pursued in ALN-VSP02 and Tekmira’s Ebola program
`which involve the inclusion of several conventional siRNAs in
`a given formulation,16 but it can also be achieved for exam-
`ple by using three- or four-stranded designs, both Dicer-
`substrate and non Dicer-substrate, in which the individual
`strands guide the cleavage of distinct targets.35 Tekmira has
`recently licensed a three-stranded RNAi trigger design from
`Halo-Bio.
`Although certainly adding to functionality, two RNAi trigger
`structures exploiting RNAi trigger structural diversity, immu-
`nostimulatory siRNAs (e.g., Alnylam)36 and single-stranded
`RNAi triggers (e.g., ISIS Pharmaceuticals)37 run counter to
`two core principles of RNAi in Man. First, it was the Nobel-
`Prize winning insight by Fire and Mello that dsRNA, and not
`for example single-stranded antisense RNA, is the trigger
`in RNAi. Second, the discovery of RNAi in mammals was
`based on the use of shorter dsRNAs that would not stimulate
`the nonspecific interferon response. It therefore remains to
`be seen whether the potency disadvantage (single-stranded
`RNAi) and safety liability (immunostimulatory siRNAs) of
`these triggers can be compensated for by their unique deliv-
`ery attributes (single-stranded RNAi) or any anticancer, anti-
`viral, or antiangiogenic effect of immunostimulatory siRNAs.
`Unlike in synthetic RNAi triggers, innovation in ddRNAi
`trigger design has somewhat stalled, particularly in the
`commercial and translational arenas, with most groups still
`employing first-generation minimal small hairpin RNAs driven
`by U6 and H1 Pol-III promoters. With safety remaining a con-
`cern for these systems,13 and the causes of toxicity still to be
`fully identified, there is considerable value to be created by
`establishing alternative ddRNAi expression systems.
`Tools to minimize RNAi-related adverse event risk. The
`challenges of drug development do not stop with hitting the
`target. In a risk-averse regulatory environment, even theo-
`rized or minor safety signals in preclinical studies can lead to
`substantial delays in the approval process. The value of tech-
`nologies that minimize adverse event risk is therefore not only
`in protecting patient safety, but also in avoiding regulatory
`surprises. In RNAi therapeutics, such technologies can be
`categorized into those that address acute toxicity and those
`that deal with the risks associated with their long-term use.
`Acute immune responses from activating innate immune
`receptors or the complement system is commonly considered
`
`Molecular Therapy–Nucleic Acids
`
`

`

`6
`
`
`RNAi Therapeutics Business 2012
`
`the next biggest scientific challenge besides delivery.8 Indeed,
`RNAi-related “flu-like symptoms” with inflammatory cytokine
`elevations have been observed in a number of clinical tri-
`als (e.g., TKM-ApoB, NucB1000, CALAA-01),12 and activa-
`tion of the alternative complement pathway was seen in the
`Atu027 trial. Although the clinical importance of such obser-
`vations can vary considerably, it is necessary to improve on
`that record. Fortunately, understanding and mitigating, if not
`abolishing acute immune responses has been one of the
`most fertile areas of RNAi therapeutics research in recent
`years. We now understand most of the relevant nucleic acid
`structural parameters (e.g., dsRNA length, GU-rich ele-
`ments) and chemical modifications (e.g., 5(cid:97)-triphosphates,
`2(cid:97)-O-methyl), as well as the innate immune receptors (TLR3,
`7, 8; RIG-I; PKR) and cell types by which such activation may
`occur. Consequently, it is possible to adjust the RNAi trigger
`structure and, in the case of synthetic RNAi triggers, apply
`chemical modifications to minimize acute immune stimula-
`tion risk, a prediction which can then be tested in predic-
`tive assays.38 While capitalizing on this progress by licensing
`related IP has proven difficult, companies like Tekmira have
`created brand value by establishing themselves as early
`adopters and experts in this area.8
`Adve

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket