throbber
C A N C E R I M M U N OT H E R A P Y
`
`Oncolytic viruses: a new class
`of immunotherapy drugs
`
`Howard L. Kaufman, Frederick J. Kohlhapp and Andrew Zloza
`
`Abstract | Oncolytic viruses represent a new class of therapeutic agents that promote
`anti-tumour responses through a dual mechanism of action that is dependent on selective
`tumour cell killing and the induction of systemic anti-tumour immunity. The molecular and
`cellular mechanisms of action are not fully elucidated but are likely to depend on viral
`replication within transformed cells, induction of primary cell death, interaction with tumour
`cell antiviral elements and initiation of innate and adaptive anti-tumour immunity. A variety
`of native and genetically modified viruses have been developed as oncolytic agents, and
`the approval of the first oncolytic virus by the US Food and Drug Administration (FDA) is
`anticipated in the near future. This Review provides a comprehensive overview of the basic
`biology supporting oncolytic viruses as cancer therapeutic agents, describes oncolytic
`viruses in advanced clinical trials and discusses the unique challenges in the development
`of oncolytic viruses as a new class of drugs for the treatment of cancer.
`
`Tumour-associated antigen
`A protein derived from tumour
`cells that can be recognized
`by the immune system.
`An immune response may be
`triggered because a T cell
`has survived negative thymic
`selection for the cognate
`antigen, or more probably
`because the normal host
`protein has been mutated
`within the cancer cell.
`
`Danger signals
`Nuclear and cytosolic proteins
`that are released by cells
`during tissue injury or necrosis,
`and stimulate the innate and
`adaptive immune system.
`
`Rutgers Cancer Institute
`of New Jersey, 195 Little
`Albany Street, Room 2004,
`New Brunswick,
`New Jersey 08901, USA.
`Correspondence to H.L.K. 
`e‑mail: howard.kaufman@
`rutgers.edu
`doi:10.1038/nrd4663
`Corrected online
`18 January 2016
`
`Oncolytic virus immunotherapy is a therapeutic approach
`to cancer treatment that utilizes native or genetically
`modified viruses that selectively replicate within tumour
`cells. The ability of viruses to kill cancer cells has been
`recognized for nearly a century, but only over the past
`decade have clinical trials documented a therapeutic
`benefit in patients with cancer1–3. Interest in oncolytic
`viruses has been increasing, based on a better under‑
`standing of viral biology, tumour immunology and
`molecular genetics. Furthermore, a recent randomized
`Phase III clinical trial demonstrated an improved durable
`response rate for patients with advanced melanoma
`who were treated with a modified herpes simplex virus
`type 1 (HSV‑1), encoding granulocyte–macrophage 
`colony‑stimulating factor (GM‑CSF) 3. This virus,
`termed talimogene laherparepvec (T‑VEC; Amgen),
`is widely anticipated to be the first oncolytic virus
`immuno therapy to be approved by the US Food and
`Drug Administration (FDA) for the treatment of can‑
`cer. The success of T‑VEC is likely to promote further
`drug development within this new class of cancer
`therapeutics.
`Of the nearly 1 million vertebrate viruses, approxi‑
`mately 320,000 are thought to infect mammalian cells4.
`Viruses have several shared properties; these include a
`genetic element composed of single‑ or double‑stranded
`DNA or RNA and the ability to infect host cells and
`replicate under permissive conditions (TABLES 1,2).
`
`The outcome of viral infections can be highly variable
`depending on the pathogenic nature of the virally
`encoded genes, interactions between the virus and the
`host immune system and the ability of the virus to repli‑
`cate and/or induce latency following infection. Insights
`into the mechanisms of viral entry, replication, induc‑
`tion and/or suppression of immune responses and lytic
`versus latent infections have led to an intense interest
`in utilizing viruses for the treatment of human diseases
`and have been used to select oncolytic vectors for the
`treatment of specific types of cancers. In contrast to
`standard viral‑based ‘vaccines’, oncolytic viruses directly
`infect and lyse tumour cells in situ. They do not nec‑
`essarily require defined antigens to be included in the
`vector as tumour-associated antigens may be released by
`dying tumour cells. Oncolytic viruses can also provide
`additional danger signals that can promote an efficient 
`antitumour immune response.
`Although incompletely understood, oncolytic viruses
`are thought to mediate antitumour activity through two
`distinct mechanisms of action: selective replication
`within neoplastic cells, resulting in a direct lytic effect
`on tumour cells; and induction of systemic antitumour
`immunity. The relative contribution of these mechanisms
`may differ depending on the nature and type of cancer
`cell, the characteristics of the viral vector, and the inter‑
`action between the virus, tumour microenvironment and
`host immune system.
`
`642 | SEPTEMBER 2015 | VOLUME 14
`
`www.nature.com/reviews/drugdisc
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 1
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Table 1 | Properties of select DNA viruses
`Adenovirus
`
`Vaccinia virus
`
`Herpesvirus
`
`Parvovirus H1
`
`35 kb
`
`190 kb
`
`154 kb
`
`5 kb
`
`70–90 nm
`
`70–100 nm
`
`200 nm
`
`18–28 nm
`
`Group I: dsDNA
`
`Adenoviridae
`
`Naked
`
`Icosahedral
`
`Group I: dsDNA
`
`Poxviridae
`
`Complex coats
`
`Complex
`
`Group I: dsDNA
`
`Herpesviridae
`
`Enveloped
`
`Icosahedral
`
`Group II: ssDNA
`
`Parvoviridae
`
`Naked
`
`Icosahedral
`
`Nucleus and cytoplasm
`
`Cytoplasm
`
`Nucleus and cytoplasm
`
`Nucleus and cytoplasm
`
`Baltimore classification
`Family
`Virion
`Capsid symmetry
`Replication site
`Cell receptor*
`Nuclear integration
`Transgene capacity
`Wild-type virus infects
`non-replicating cells
`Virulence of wild-type virus‡
`Antivirals
`Immunogenicity¶
`Haemagglutination
`Blood–brain barrier
`penetration
`Achievable titre (PFU per ml)
`5 × 108
`1010
`109
`1012
`CAR, coxsackie-adenovirus receptor; dsDNA, double-stranded DNA; HVEM, herpesvirus entry mediator; MTD, maximum tolerated dose; N/A, not applicable;
`PFU, plaque forming unit; ssDNA, single-stranded DNA. *Only well-validated receptors included, others may have been reported. ‡In humans. ¶Upon re-exposure.
`
`CAR
`
`+
`
`++
`
`–
`
`+/–
`
`+
`
`–
`
`+/–
`
`–
`
`Unknown
`
`HVEM, nectin 1, nectin 2
`
`Sialic acid residues
`
`–
`
`+++
`
`–
`
`+/–
`
`+
`
`–
`
`–
`
`–
`
`+
`
`+++
`
`–
`
`–
`
`+
`
`–
`
`–
`
`–
`
`+
`
`N/A
`
`+
`
`+
`
`–
`
`+
`
`+
`
`+
`
`Certain viruses have the ability to enter cancer cells
`and selectively replicate within such cells. Although
`oncolytic viruses can enter both normal and cancer cells,
`the inherent abnormalities in the cancer cell response to
`stress, cell signalling and homeostasis provide a selective
`advantage for viral replication5. The normal host cell anti‑
`viral machinery, which is responsible for the detection and
`clearance of viruses, may also be abnormal in cancer cells.
`For example, the protein kinase R (PKR) is a critical factor
`that helps in clearing intracellular viral infections. PKR
`may be absent in some cancer cells, allowing increased
`viral replication, whereas it may be active in other cancer
`cells, such as low‑grade tumours, and these differences
`can influence the therapeutic activity of an oncolytic virus.
`The immune response to oncolytic viruses appears to
`be an important component of the antitumour effect, but
`it can be a double‑edged sword. On the one hand, viruses
`can help to promote an immune response against the
`tumour cells by allowing tumour antigen presentation
`in the context of an active viral infection. On the other
`hand, neutralizing antiviral responses may block virus
`replication and ongoing infection of tumour cells. The
`therapeutic outcome depends on a complex interplay
`
`between these opposing forces. When systemic immunity
`is fully engaged, however, therapeutic responses may be
`seen in both locally injected tumours and at distant sites
`of un‑infected tumour growth.
`Many viruses have been proposed as vectors for
`oncolytic virus immunotherapy, and considerable work
`has been done to optimize viral vectors by attenuating
`pathogenicity and enhancing immunogenicity6. To date,
`adenoviruses, poxviruses, HSV‑1, coxsackieviruses,
`poliovirus, measles virus, Newcastle disease virus
`(NDV), reovirus, and others, have entered into early‑
`phase clinical trials. Two viruses, T‑VEC and H101
`(Shanghai Sunway Biotech) have now achieved regula‑
`tory review. H101 is a genetically modified oncolytic
`adenovirus that, in combination with chemotherapy,
`was approved for the treatment of nasopharyngeal
`carcinoma in China in November 2005 (REFS 3,7).
`This Review provides a comprehensive overview
`of critical issues in the development of oncolytic virus
`immunotherapy. We discuss preclinical and clinical data
`that support a role for oncolytic viruses in cancer therapy
`and detail some of the unique challenges in oncolytic
`viruse drug development.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 14 | SEPTEMBER 2015 | 643
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 2
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Table 2 | Properties of select RNA viruses
`Reovirus
`Coxsackievirus
`
`Seneca
`Valley Virus
`
`Poliovirus
`
`Measles virus
`
`Newcastle
`disease virus
`
`Vesicular
`stomatitis
`virus
`
`23 kb
`
`28 kb
`
`7 kb
`
`7.5 kb
`
`16 kb
`
`15 kb
`
`11 kb
`
`75 nm
`
`28 nm
`
`25–30 nm
`
`30 nm
`
`100–200 nm
`
`100–500 nm
`
`80 nm
`
`Baltimore
`classification
`
`Group III:
`dsRNA
`
`Group IV: ssRNA
`
`Group IV: ss(+)
`RNA
`
`Group IV: ss(+)
`RNA
`
`Group V: ss(–)
`RNA
`
`Group V: ss(–)
`RNA
`
`Group V ss(–)
`RNA
`
`Family
`Virion
`Capsid symmetry
`Replication site
`Cell receptor*
`
`Reoviridae
`
`Picornaviridae
`
`Picornaviridae
`
`Picornaviridae
`
`Paramyxoviridae Paramyxoviridae
`
`Rhabdoviridae
`
`Naked
`
`Naked
`
`Naked
`
`Naked
`
`Enveloped
`
`Enveloped
`
`Enveloped
`
`Icosahedral
`
`Icosahedral
`
`Icosahedral
`
`Icosahedral
`
`Icosahedral
`
`Helical
`
`Helical
`
`Cytoplasm Cytoplasm
`
`Cytoplasm
`
`Cytoplasm
`
`Cytoplasm
`
`Cytoplasm
`
`Cytoplasm
`
`Unknown
`
`Unknown
`
`CD155
`
`SLAM and CD46 Unknown
`
`LDLR
`
`–
`
`–
`
`CAR/ICAM-1/
`DAF
`–
`
`N/A
`
`–
`
`+/–
`
`–
`
`–
`
`+
`
`–
`
`–
`
`N/A
`
`+
`
`+
`
`–
`
`+
`
`+
`
`+
`
`+
`
`–
`
`109
`
`109
`
`N/A
`
`Nuclear integration –
`Transgene capacity N/A
`Wild-type virus
`+
`infects non-
`replicating cells
`Virulence of wild-
`type virus‡
`Antivirals
`Immunogenicity¶
`–
`Haemagglutination +
`Blood–brain barrier
`+
`penetration
`Achievable titre
`(PFU per ml)
`CAR, coxsackie-adenovirus receptor; DAF, decay accelerating factor; dsRNA, double-stranded RNA; ICAM-1, intercellular adhesion molecule 1; LDLR, low-density
`lipoprotein receptor; MTD, maximum tolerated dose; N/A, not applicable; PFU, plaque forming unit; SLAM, signalling lymphocytic activation molecule; ss(+)RNA,
`positive single-stranded RNA; ss(–)RNA, negative single-stranded RNA; VP, viral particle. *Only well-validated receptors included, others may have been reported.
`‡To humans. ¶Upon re-exposure.
`
`–
`
`N/A
`
`–
`
`–
`
`–
`
`+/–
`
`+
`
`+
`
`108
`
`–
`
`+
`
`–
`
`–
`
`–
`
`–
`
`–
`
`–
`
`+
`
`–
`
`+
`
`–
`
`–
`
`–
`
`+
`
`+
`
`+
`
`+
`
`–
`
`–
`
`–
`
`–
`
`1011
`
`108
`
`2 × 1010
`
`Cell antiviral response
`elements
`Intracellular and extracellular
`components of the inherent
`cellular response to viral
`infection, including detection
`(via protein kinase R (PKR),
`Toll-like receptors (TLRs),
`retinoic acid-inducible gene 1
`(RIG-1), and others) and
`initiation of the immune
`response through the release
`of cytokines (such as type I
`interferons), danger-associated
`molecular pattern signals
`(DAMPs) (such as high mobility
`group box 1(HMGB1),
`heat shock proteins (HSPs),
`and others), and pathogen-
`associated molecular patterns
`(PAMPs; viral products
`recognized by TLRs).
`
`Mechanisms of oncolytic virus immunotherapy
`The mechanisms through which oncolytic viruses mediate
`tumour rejection are incompletely understood. Most
`oncolytic viruses directly kill host tumour cells. This
`activity is influenced by the efficiency of cell receptor
`targeting, viral replication and host cell antiviral response
`elements8,9. The lytic potential of oncolytic viruses also
`depends on the type of virus, dose, natural and induced
`viral tropism, and the susceptibility of the cancer cell
`to the different forms of cell death (apoptosis, necrosis,
`pyroptosis and autophagy).
`In normal cells, a variety of signalling pathways oper‑
`ate to detect and clear pathogenic viral particles (FIG. 1).
`These pathways can be stimulated by local interferon
`(IFN) release or through intracellular Toll‑like receptors
`(TLRs), which are activated by viral elements. TLRs are cell
`surface and intracellular pattern recognition receptors that
`are activated in response to repeated sequences, so‑called
`pathogen‑associated molecular patterns (PAMPs), which
`
`are common to pathogenic bacteria and viruses. PAMPs
`may include elements of viral capsids, DNA, RNA and
`viral protein products. TLR signalling activates host
`cell antiviral responses and systemic innate immunity.
`Several downstream host cell factors involved in onco‑
`lytic virus clearance have been identified, including TNF‑
`associated factor 3 (TRAF3), IFN‑related factor 3 (IRF3),
`IRF7 and retinoic acid‑inducible gene 1 (RIG‑1). These
`factors activate the JAK–STAT (Janus kinase–signal
`transducer and activator of transcription) pathway, which
`coordinates the antiviral machinery in infected cells. The
`antiviral machinery reinforces local IFN release, which
`activates PKR activity. PKR is an intracellular protein
`kinase that recognizes double‑stranded RNA and other
`viral elements10,11. When activated by viral elements, PKR
`terminates cell protein synthesis and promotes rapid cell
`death and viral clearance. In cancer cells, IFN pathway
`signalling and PKR activity may be abnormal, and viral
`clearance is thwarted12.
`
`644 | SEPTEMBER 2015 | VOLUME 14
`
`www.nature.com/reviews/drugdisc
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 3
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`a Healthy cell
`
`b Cancer cell
`
`IFN
`
`IFNR
`
`IFN
`
`IFNR
`
`Viral TLR
`ligands
`
`JAK
`
`Cytoplasm
`
`TLR
`
`Viral TLR
`ligands
`
`TLR
`
`࢑
`
`JAK
`
`PKR
`
`(cid:127) Reovirus
`(cid:127) Herpes
`(cid:127) Adenovirus
`(cid:127) Vaccinia
`(cid:127) Influenza
`
`Translation
`
`dsRNA
`
`MYD88
`
`TRIF
`
`࢑
`
`RIG-1
`NDV
`
`TRAF6
`
`TRAF3
`
`dsRNA
`
`MYD88
`
`TRIF
`
`RIG-1
`NDV
`
`TRAF6
`
`TRAF3
`
`NF-κB
`
`IRF7
`NDV
`
`IRF3
`NDV
`
`STAT
`
`IRF9
`
`(cid:127) VSV
`(cid:127) NDV
`(cid:127) Measles
`(cid:127) Vaccinia
`
`Apoptosis
`
`NF-κB
`
`Nucleus
`
`࢑
`IRF7
`NDV
`
`࢑
`
`IRF3
`NDV
`
`࢑
`
`PKR
`
`(cid:127) Reovirus
`(cid:127) Herpes
`(cid:127) Adenovirus
`(cid:127) Vaccinia
`(cid:127) Influenza
`
`STAT
`
`࢑
`IRF9
`
`(cid:127) VSV
`(cid:127) NDV
`(cid:127) Measles
`(cid:127) Vaccinia
`
`Translation
`
`Apoptosis
`
`Pro-
`inflammatory
`cytokines
`
`NF-κB
`
`Type I
`IFNs
`
`IRF7
`
`IRF3
`
`(cid:127) IRF7
`(cid:127) PKR
`
`STAT
`
`IRF9
`
`Pro-
`inflammatory
`cytokines
`
`NF-κB
`
`Type I
`IFNs
`
`IRF7
`
`IRF3
`
`(cid:127) IRF7
`(cid:127) PKR
`
`STAT
`
`IRF9
`
`Figure 1 | Oncolytic viruses can exploit cancer immune evasion pathways. a | Following viral infection, most
`normal cells activate an antiviral pathway that allows to contain viral infections. The antiviral machinery can be
`triggered by viral pathogen-associated molecular patterns (PAMPs) that activate Toll-like receptors (TLRs) or through
`the detection of viral nucleic acids by retinoic acid-inducible gene 1 (RIG-1). Once a virus is detected, a signalling
`Nature Reviews | Drug Discovery
`cascade through several type I interferon (IFN) elements (Janus kinase (JAK), signal transducer and activator of
`transcription (STAT), and interferon regulatory factor 9 (IRF9)) results in a programmed transcriptional pathway that
`limits viral spread and can target infected cells for apoptosis or necrosis. Local IFN production induced by the innate
`immune response to viral infections may also promote antiviral activity through the IFN receptor (IFNR). TLRs signal via
`the myeloid differentiation primary response protein MYD88, TIR-domain-containing adapter-inducing IFNβ (TRIF),
`IRF7, IRF3 and nuclear factor-κB (NF-κB), inducing the production of pro-inflammatory cytokines and type I IFNs.
`The type I IFNs signal through the JAK–STAT signalling pathway, resulting in the upregulation of cell cycle regulators,
`such as protein kinase R (PKR) and IRF7, which limit viral spread by binding to viral particles and triggering type I IFN
`transcriptional pathways, promoting abortive apoptosis of infected cells and the production of cytokines that alert the
`immune system to the presence of a viral infection. b | In cancer cells, however, this process is disrupted. Cancer cells
`may downregulate key signalling components within the innate signalling pathway, including RIG-1, IRF7, and IRF3
`(REF. 1). This limits detection of viral particles by TLR and RIG-1, making cancer cells more susceptible to viral
`replication. Furthermore, cancer cells may downregulate key components of the type I IFN signalling pathway2–7,
`thereby limiting the pro-apoptotic and cell cycle regulatory effects of type I IFNs. Although data are limited, the figure
`depicts individual viruses near the factors and/or pathways that are known to promote viral elimination in normal cells
`(part a) or that support viral replication owing to factor deficiency in cancer cells (part b). dsRNA, double-stranded
`RNA; NDV, Newcastle disease virus; TRAF, TNF-associated factor; VSV, vesicular stomatitis virus.
`
`Different viruses can also manipulate distinct aberrant
`signalling factors within tumour cells to block apoptosis,
`which allows more time for the virus to complete its life
`cycle. Following viral replication, most oncolytic viruses
`induce cell death, which can directly eliminate viable
`tumour cells but also sets the stage for initiating systemic
`immune responses. Induction of host immune responses
`can be greatly aided by both the type of cell death and the
`
`release of danger signals from virus‑infected cells. For
`example, necrosis or pyroptosis are more immunogenic
`forms of cell death than apoptosis.
`
`Induction of systemic anti-tumour immunity. The induction
`of systemic innate and tumour‑specific adaptive immune
`responses appears to be a critical element for tumour
`eradication with oncolytic viruses. Following oncolytic
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 14 | SEPTEMBER 2015 | 645
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 4
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`cell death, tumour cells release tumour‑associated
`antigens that can serve to promote an adaptive immune
`response that mediates tumour regression at distant
`tumour sites that are not exposed to virus. They also
`release viral PAMPS and additional cellular danger‑
`associated molecular pattern signals (DAMPs; for
`example, heat shock proteins, high mobility group box 1
`(HMGB1) protein, calreticulin, ATP, and uric acid) and
`cytokines (for example, type I IFNs, tumour necrosis
`factor‑α (TNFα), IFNγ, and interleukin‑12 (IL‑12)), which
`promote the maturation of antigen‑presenting cells (APCs)
`such as dendritic cells. These activate antigen‑specific CD4+
`and CD8+ T cell responses. Once activated, CD8+ T cells
`can expand into cytotoxic effector cells with the ability to
`traffic to sites of established tumour growth, where they
`mediate anti‑tumour immunity upon antigen recognition
`(FIG. 2). However, the natural ability of viruses to induce a
`host antiviral immune response may result in clearance of
`the virus through neutralizing antiviral antibodies and/or
`cytotoxic T‑cell‑mediated immune responses. The extent
`to which viral neutralization influences the induction of
`tumour immunity is complex and can be influenced by
`many variables, most notably the characteristics of the
`virus and the tumour microenvironment.
`The release of tumour‑associated antigens, especially in
`combination with local cytokine and DAMP release, can
`be beneficial for inducing innate and adaptive immune
`responses against cancer cells (FIG. 2). This effect may be
`especially important for mediating tumour regression at
`distant tumour sites that are not injected or exposed to
`virus. Preclinical studies have demonstrated the impor‑
`tance of tumour‑specific CD8+ T cells in mediating tumour
`rejection with oncolytic viruses13.
`Type I IFNs and DAMPs can also directly activate
`natural killer (NK) cells, which are part of the innate
`immune response. NK cells can kill target cells with
`downregulated major histocompatibility complex (MHC)
`class I expression, which is a common occurrence in
`cancer cells14,15. The influence of NK cells may depend
`on both the host species and the characteristics of the
`virus16,17. Furthermore, NK cells may be detrimental to
`the effectiveness of oncolytic viruses by eliminating virally
`infected cells18. The factors that influence the balance
`between immune‑mediated viral clearance and induction
`of antitumour immunity are incompletely understood.
`
`Counteracting cancer-mediated immune evasion. Cancer
`cells have evolved sophisticated strategies for avoiding
`immune‑mediated destruction. For example, tumour
`cells and the microenvironment can express immune‑
`inhibitory surface receptors that inactivate effector
`immune cells, and secrete factors — such as IL‑10, trans‑
`forming growth factor‑β (TGFβ) and indoleamine‑2,3‑
`dioxygenase (IDO) — that facilitate the recruitment of
`immune‑suppressive cells, such as tumour‑associated
`macrophages19 and myeloid‑derived suppressor cells20,
`to sites of tumour growth21. Oncolytic viruses modify
`this suppressive microenvironment through a variety
`of mechanisms that alter the cytokine milieu and the
`type of immune cells within the tumour microenviron‑
`ment22,23. These changes promote immune‑mediated
`
`tumour cell recognition and eradication, and can trigger
`tumour‑associated antigen and epitope spreading24,25.
`In the presence of danger signals and TLR engagement,
`the levels of type I IFNs and other inflammatory media‑
`tors increase, further potentiating systemic immunity
`against the cancer.
`The killing of cancer cells can result in the release of
`novel cancer antigens (neo-antigens) that may have been
`previously hidden to the immune system because of
`restricted presentation (FIG. 2). This effect was recently
`reported following the treatment of cancer patients with
`immunotherapeutic T cell checkpoint inhibitors26,27. Such
`neo‑antigens may be taken up by local APCs in the context
`of a pro‑inflammatory environment, which can trigger an
`immune response against the neo‑antigen. If new T cell
`clones are generated, they may be able to circulate and kill
`antigen‑expressing cancer cells, including cancer cells that
`were not infected by the virus. The immune response has
`also been associated with a ‘immune‑associated’ bystander
`effect, in which local release of cytotoxic perforins and
`granzymes may result in the killing of nearby tumour cells,
`even in the absence of direct antigen expression28. This
`is distinct from the virus bystander effect (BOX 1), which
`relates to the replication of the virus inside cancer cells and
`its spread to previously un‑infected cancer cells.
`
`Oncolytic virus biodistribution. Physical barriers that
`reduce the spread of oncolytic viruses include necrosis,
`calcification, hypoxia, acidosis, increased proteolytic
`activity, and a high interstitial pressure29–31. Furthermore,
`tumours are dense with extracellular matrix and are
`poorly vascularized. The majority of clinical studies
`with oncolytic viruses (such as adenovirus, poxvirus,
`HSV‑1, measles, and reovirus) have used intratumoural
`injections to bypass the tumour architectural barriers.
`However, intratumoural injections are limited to tumours
`that are physically accessible through clinical palpation or
`direct imaging. As discussed above, injection of an onco‑
`lytic virus into one tumour lesion can induce a systemic
`anti‑tumour response that can overcome physical limita‑
`tions, as evidenced in the T‑VEC OPTIM Phase III clini‑
`cal trial3. Other viruses, such as Seneca Valley Virus, can
`be delivered intravenously because of natural resistance
`to haemagglutination, a process resulting in premature
`viral clearance and reduced delivery to the tumour site
`following intravenous delivery32.
`The blood–brain barrier may limit the ability of some
`viruses (and many other drugs) to reach primary brain
`tumours and brain metastases. This may be overcome
`by direct injection into central nervous system (CNS)
`tumours or through the use of external reservoirs that
`communicate with sites of brain tumours. Parvovirus
`naturally crosses the blood–brain barrier, allowing for the
`delivery of this oncolytic virus via the intravascular route.
`Parvovirus H‑1PV has been used in clinical trials of glio‑
`blastoma multiforme (GBM)33. However, there have been
`relatively few studies of oncolytic virus distribution in vivo
`to evaluate viral penetrance throughout the CNS33–35.
`Tumour size and heterogeneity can present another
`barrier to virus biodistribution. Moreover, growth‑
`arrested cancer cells in hypoxic environments are less
`
`Epitope spreading
`A process whereby tissue
`damage during an initial
`immune response against
`one antigen may lead to the
`priming of self-reactive T
`and/or B cells targeting other
`areas (or epitopes) within
`the initial antigen or against
`other antigens.
`
`Neo‑antigens
`New antigens, often derived
`from cell metabolic pathway
`proteins, and commonly
`expressed in tumour cells.
`Neo-antigens may appear
`as a consequence of epitope
`spreading following initial
`immune attack on a tumour
`cell triggered by an unrelated
`antigen.
`
`Checkpoint inhibitors
`Monoclonal antibodies that
`inhibit negative immune
`checkpoints. Immune
`checkpoints control important
`intracellular signalling
`pathways in the immune
`system that either activate
`or inhibit immune responses.
`Checkpoint inhibitors have
`shown significant promise in
`the treatment of cancer.
`Most notably, blockade of
`cytotoxic T lymphocyte
`antigen 4 (CTLA4) and
`interactions between
`programmed cell death 1
`(PD1) and programmed cell
`death 1 ligand 1 (PDL1) have
`had significant impact on
`the treatment of melanoma,
`lung cancer, and possibly
`many other cancers.
`
`646 | SEPTEMBER 2015 | VOLUME 14
`
`www.nature.com/reviews/drugdisc
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 5
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Oncolytic
`virus
`
`Infection
`
`Cancer cell
`
`(cid:127) Viral proteins
`(cid:127) Viral genome
`
`Release/
`secrete
`
`(cid:127) ER stress
`(cid:127) Genotoxic stress
`
`ROS
`
`Cytokine
`receptors
`
`NK cell
`
`Cytotoxicity
`
`CD8+ T cell
`
`Cytokine
`receptors
`
`Cytotoxicity
`
`IL-2R
`
`IL-2
`
`Viral
`oncolysis
`
`ROS
`
`Release
`
`Release
`
`CD28
`
`TCR
`
`CD4+ T cell
`
`Antigen
`uptake
`
`TLR
`
`MHC MHC
`
`CD40L
`
`PAMPs
`(cid:127) Viral capsids
`(cid:127) Viral DNA
`(cid:127) Viral dsRNA/ssRNA
`(cid:127) Viral proteins
`
`DAMPs
`(cid:127) HSPs
`(cid:127) HMGB1
`(cid:127) Calreticulin
`(cid:127) ATP
`(cid:127) Uric acid
`
`Cytokines
`(cid:127) Type I interferons
`(cid:127) TNFα
`(cid:127) IFNγ
`(cid:127) IL-12
`
`(cid:127) Type I IFNs
`(cid:127) DAMPs/PAMPs
`
`(cid:127) Type I IFNs
`(cid:127) Cytokines
`
`DAMPs/
`PAMPs
`
`(cid:127) Viral antigens
`(cid:127) TAAs/neoantigens
`
`(cid:127) CD80/CD86
`(cid:127) Chemokine receptors
`
`Activation
`
`CD40
`
`TCR
`
`MHC
`
`Antigen presenting cell
`
`Figure 2 | The induction of local and systemic anti-tumour immunity by oncolytic viruses. The therapeutic efficacy
`of oncolytic viruses is determined by a combination of direct cancer cell lysis and indirect activation of anti‑tumour
`immune responses. Upon infection with an oncolytic virus, cancer cells initiate an antiviral response that consists of
`Nature Reviews | Drug Discovery
`endoplasmic reticulum (ER) and genotoxic stress. This response leads to the upregulation of reactive oxygen species (ROS)
`and the initiation of antiviral cytokine production. ROS and cytokines, specifically type I interferons (IFNs), are released
`from the infected cancer cell and stimulate immune cells (antigen presenting cells, CD8+ T cells, and natural killer (NK)
`cells). Subsequently, the oncolytic virus causes oncolysis, which releases viral progeny, pathogen-associated molecular
`patterns (PAMPs), danger-associated molecular pattern signals (DAMPs), and tumour associated antigens (TAAs)
`including neo-antigens. The release of viral progeny propagates the infection with the oncolytic virus. The PAMPs
`(consisting of viral particles) and DAMPs (comprising host cell proteins) stimulate the immune system by triggering
`activating receptors such as Toll-like receptors (TLRs). In the context of the resulting immune‑stimulatory environment,
`TAAs and neo-antigens are released and taken up by antigen presenting cells. Collectively, these events result in the
`generation of immune responses against virally infected cancer cells, as well as de novo immune responses against
`TAAs/neo-antigens displayed on un‑infected cancer cells. CD40L, CD40 ligand; dsRNA, double-stranded RNA;
`HMGB1, high mobility group box 1; HSP, heat shock protein; IL-2, interleukin-2; IL-2R, IL-2 receptor; MHC, major
`histocompatibility complex; ssRNA, single-stranded RNA; TCR, T cell receptor; TNFα, tumour necrosis factor-α.
`
`likely to be permissive to infection31,36,37. Stromal cells,
`such as cancer‑associated fibroblasts, may be infected
`by oncolytic viruses but are non‑permissive to viral
`replication. Thus, fibroblasts may act as a decoy reservoir
`for oncolytic viruses, reducing the delivery of infectious
`virions to cancer cells38. Another mechanism that may
`limit the overall effectiveness of oncolytic viruses is the
`susceptibility of cancer cells to apoptosis, which may
`be induced by viral infection or other factors39. If cells
`undergo apoptosis too rapidly, this will reduce the time
`
`for viral replication and propagation and decrease the
`amount of active virus in the tumour, ultimately limiting
`the active intratumoural dose.
`
`Development of oncolytic viruses as drugs
`As oncolytic viruses are live viral particles, the over‑
`all design of oncolytic virus strategies must consider
`approaches to tumour cell targeting and attenuating
`viral pathogenesis, as well as approaches to limit viral
`immunogenicity while promoting tumour cell killing
`
`NATURE REVIEWS | DRUG DISCOVERY
`
` VOLUME 14 | SEPTEMBER 2015 | 647
`
`© 2016 Macmillan Publishers Limited. All rights reserved
`
`R E V I E W S
`
`Replimune Limited Ex. 2014 - Page 6
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Box 1 | Virus bystander effect
`
`The optimal method for the administration of oncolytic viruses is not established but
`most clinical trials have utilized direct injection into established tumours, with some
`evaluating intravenous delivery. Because oncolytic viruses are capable of replication,
`only a limited number of tumour cells need to be infected. A significant bystander effect
`can be anticipated by the local replication of the virus and its release into the
`surrounding tumour where viral particles can then infect new tumour cells. If the host
`antiviral immune response does not neutralize the virus, infection may continue to
`propagate. The inclusion of suicide genes within the oncolytic virus can further enhance
`the bystander effect and achieve much greater cell killing. However, the contribution
`of the bystander effect to the overall therapeutic effectiveness of the oncolytic virus
`needs to be demonstrated in a randomized clinical trial to be more fully evaluated.
`
`and immunogenicity. The flexibility of recombinant
`engineering has allowed the exploration of a number of
`strategies to enhance the effectiveness of oncolytic viruses.
`
`Targeting oncolytic viruses to cancer cells. Many of the
`oncolytic viruses that are currently in the clinic have a
`natural tropism for cell surface proteins that are aber‑
`rantly expressed by cancer cells (FIG. 3). For example,
`HSV‑1 uses the herpesvirus entry mediator (HVEM) and
`selected nectins for cell entry. These surface receptors are
`overexpressed on some cancer cells, including melanoma
`and various carcinomas40. Measles virus, specifically the
`Edmonston strain, utilizes the surface receptor CD46
`for cell entry41. CD46 normally func

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