throbber
Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 1 of 22 PageID #: 12578
`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 1 of 22 PagelD #: 12578
`
`EXHIBIT 5
`EXHIBIT 5
`
`

`

`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 2 of 22 PageID #: 12579
`Article
`
`SARS-CoV-2 mRNAvaccine design enabled
`by prototype pathogen preparedness
`
`
`
`https://doi.org/10.1038/s41586-020-2622-0
`Received: 10 June 2020
`
`
`Accepted: 29 July 2020
`
`Published online: 5 August 2020
`
`|® Check for updates
`
` Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 2 of 22 PagelD #: 12579
`
`Since its emergence in December 2019, SARS-CoV-2 has accounted for
`more than 30 million cases of coronavirus disease 2019 (COVID-19)
`worldwide in 9 months?. SARS-CoV-2is the third novel Betacoronavirus
`in the past 20 yearsto cause substantial human disease; however, unlike
`its predecessors SARS-CoVand Middle East respiratory syndrome
`coronavirus (MERS-CoV), SARS-CoV-2 is transmitted efficiently from
`person to person.In the absenceof a vaccine, public health measures
`such as quarantine of newly diagnosed cases, contacttracing, use of
`face masks and physical distancing have been putinto place toreduce
`transmission‘. It is estimated that until 60-70% of the population have
`immunity, COVID-19 is unlikely to be sufficiently well-controlled for
`normal human activities to resume.Ifimmunity remains solely depend-
`ent oninfection, even at acasefatality rate of1%, more than4O million
`people could succumb to COVID-19 globally*. Therefore, rapid develop-
`mentof vaccines against SARS-CoV-2will be critical for changing the
`global dynamicsofthis virus.
`The spike (S) protein, a class I fusion glycoprotein analogous to
`influenza haemagglutinin, respiratory syncytial virus (RSV) fusion
`glycoprotein (F) and human immunodeficiency virus gp160 (Env),
`
`Kizzmekia S. Corbett", Darin K. Edwards”, Sarah R. Leist*"°, Olubukola M. Abiona’,
`Seyhan Boyoglu-Barnum', RebeccaA.Gillespie’, Sunny Himansu?, Alexandra Schafer’,
`Cynthia T. Ziwawo', AnthonyT. DiPiazza', Kenneth H. Dinnon®, Sayda M.Elbashir’,
`Christine A. Shaw’, Angela Woods’, EthanJ. Fritch*, David R. Martinez®, Kevin W. Bock°,
`Mahnaz Minai’, Bianca M. Nagata’, Geoffrey B. Hutchinson’, Kai Wu’, Carole Henry’, Kapil Bahl’,
`Dario Garcia-Dominguez’, LingZhi Ma’, Isabella Renzi”, Wing-Pui Kong’, Stephen D. Schmidt',
`Lingshu Wang, YiZhang', Emily Phung", Lauren A. Chang’, RebeccaJ. Loomis’,
`Nedim Emil Altaras?, Elisabeth Narayanan’,Mihir Metkar?, Vlad Presnyak?, Cuiping Liu’,
`MarkK. Louder’, Wei Shi’, Kwanyee Leung’, Eun Sung Yang’, Ande West®, Kendra L. Gully’,
`Laura J. Stevens’, Nianshuang Wang*, Daniel Wrapp*,Nicole A. Doria-Rose’,
`Guillaume Stewart-Jones?, Hamilton Bennett’, Gabriela S. Alvarado’, Martha C. Nason’,
`Tracy J. Ruckwardt', Jason S. McLellan®, Mark R. Denison’, James D. Chappell’, lan N. Moore®,
`Kaitlyn M. Morabito’, John R. Mascola’, Ralph S. Baric**, Andrea Carfi?™ & Barney S. Graham'™
`
`Avaccinefor severe acute respiratory syndrome coronavirus2 (SARS-CoV-2)is
`needed to control the coronavirus disease 2019 (COVID-19) global pandemic.
`Structural studies have led to the developmentof mutationsthatstabilize
`Betacoronavirus spike proteinsin the prefusionstate, improving their expression and
`increasing immunogenicity’. This principle has been applied to design mRNA-1273, an
`mRNAvaccine that encodesa SARS-CoV-2 spike protein thatis stabilized in the
`prefusion conformation. Here we show that mRNA-1273 inducespotentneutralizing
`antibody responsesto both wild-type (D614) and D614G mutant? SARS-CoV-2 as well
`as CD8* T cell responses, and protects against SARS-CoV-2 infection in the lungs and
`nosesof mice without evidence of immunopathology. mRNA-1273is currently ina
`phaseIll trial to evaluateitsefficacy.
`
`is the major surface protein on the coronavirus virion and thepri-
`marytarget for neutralizing antibodies. S proteins undergo marked
`structural rearrangementto fuse virus and host cell membranes,
`enabling delivery of the viral genome into targetcells. We previously
`showedthat prefusion-stabilized protein immunogens that preserve
`neutralization-sensitive epitopesare an effective vaccine strategy
`for enveloped viruses such as RSV°”. Subsequently,we identified 2
`proline substitutions (2P) at the apex of the central helix and heptad
`repeat 1 that effectively stabilized MERS-CoV, SARS-CoV and human
`coronavirus HKU1S proteinsinthe prefusion conformation“ Similar
`to other prefusion-stabilized fusion proteins, MERS-CoV S(2P) protein
`was more immunogenic at lower doses than wild-type S protein’. The
`2P mutationhas similar effects on the stability ofS proteins from other
`betacoronaviruses, suggesting a generalizable approachfordesigning
`stabilized-prefusion BetacoronavirusS protein antigensfor vaccina-
`tion. Such generalizability is fundamental to the prototype pathogen
`approachfor pandemic preparedness®*.
`Coronaviruses have long been predicted to havea high probability of
`causing zoonotic disease and pandemics’.As part ofour pandemic
`
`‘Vaccine Research Center, NationalInstitute of Allergy and Infectious Diseases, NationalInstitutes of Health, Bethesda, MD, USA. Moderna Inc, Cambridge, MA, USA. "Department of
`Epidemiology, Gillings Schoolof Global Public Health, University of North Carolina at ChapelHill, Chapel Hill, NC, USA. ‘Departmentof Microbiology and Immunology, School of Medicine,
`University of North Carolina at ChapelHill, ChapelHill, NC, USA. °NationalInstitute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.“institute for Biomedical
`Sciences, George Washington University, Washington, DC, USA. Departmentof Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA. "Departmentof Molecular Biosciences,
`University of Texas at Austin, Austin, TX, USA. °Biostatistics Research Branch,Division ofClinical Research, NationalInstitute of Allergy and Infectious Diseases, NationalInstitutes of Health,
`Bethesda, MD, USA. These authorscontributed equally: Kizzmekia S. Corbett, Darin K. Edwards, Sarah R. Leist. e-mail: andrea.carfi@modernatx.com; bgraham@nih.gov
`
`Nature | Vol586 | 22 October 2020 | 567
`
`

`

`andcorrelates of protection. Achieving an effective and rapid vac-
`@=~Neutralizing antibodies 9oa
`cine response to a newly emerging virus requires both the precision
`z 6
`g 110
`ie PES
`5
`~e 0.01 pg
`afforded by structure-based antigen design and a manufacturing
`ie 7
`8 100
`eas
`platform to shorten time to productavailability. Producingcell lines
`= 4
`~~ 1g
`8
`2 00
`and clinical-grade subunit protein typically takes more than one year,
`3
`i
`whereas manufacturing nucleic acid vaccines can be achieved ina mat-
`. a
`2
`ter of weeks””"®, In addition to advantagesin manufacturing speed,
`2 70
`mRNAvaccinesare potently immunogenic and elicit both humoral
`& 901234567 8 9101112
`and cellular immunity’. We therefore evaluated mRNA formulated
`Time after challenge (d)
`inlipid nanoparticles (mRNA-LNP)asa delivery vehicle for MERS-CoV
`S(2P), and found that transmembrane-anchored MERS-CoVS(2P) mRNA
`elicited more potent pseudovirus-neutralizing antibody responses
`than secreted MERS-CoVS(2P) (Extended DataFig. 1a). Additionally,
`consistent with protein immunogens, MERS-CoV S(2P) mRNA was
`more immunogenic than wild-type MERS-CoVS mRNA(Extended Data
`Fig. 1b). Immunization with MERS-CoV S(2P) mRNA-LNP elicited potent
`pseudovirus-neutralizing activity with a dose as lowas0.1 pg and pro-
`tected transgenic mice expressing human DPP4(288/330*")” against
`lethal MERS-CoV challenge ina dose-dependentmanner,establishing
`that mRNA encoding S(2P)proteinis protective. Notably, a subprotec-
`tive 0.01,1g dose of MERS-CoV S(2P) mRNAdid not cause exaggerated
`disease following MERS-CoVinfection, but instead resultedin par-
`tial protection against weight loss followed byfull recovery without
`Fig.1| MERS-CoVS-2P mRNAprotectsmice from lethal challenge.
`evidenceof enhancedillness (Fig. 1).
`a-d, 288/330" mice were immunized at weeks 0 and3with0.01 (green), 0.1
`SARS-CoV-2 wasfirst identified as the cause of an outbreak of res-
`(blue) or 1 pg (red) MERS-CoV S(2P) mRNA.Control mice wereadministered
`piratory disease in Wuhan, Chinain earlyJanuary 2020. Within 24 h of
`phosphate-bufferedsaline (PBS) (grey).a, Two weekspost-boost, sera were
`the release of genomic sequences of SARS-CoV-2isolates on 10Janu-
`collected fromthree mice per group and assessed for neutralizing antibodies
`ary 2020, the 2P mutations were substitutedinto S protein residues
`against MERS m35c4 pseudovirus. b-d, Four weeks post-boost, 12 mice per
`986 and 987 to produce prefusion-stabilized SARS-CoV-2 S(2P) pro-
`groupwerechallenged witha lethal dose ofmouse-adapted MERS-CoV
`tein for structural analysis” and serological assay development”**>
`(m35c4). b, Following challenge, mice were monitoredfor weight loss. c,d, Two
`dayspost-challenge,at peak viral load,lungviral titres (c) and haemorrhage
`in silico, without additional experimentalvalidation. Within 5 days
`(scored as: 0,no haemorrhage, 4, severe haemorrhageinall lobes) (d) were
`ofthe release of the sequence, current good manufacturing practice
`assessed from five mice per group.Inc,d, alldoselevels were compared by
`(cGMP) production of mRNA-LNPencoding the SARS-CoV-2 S(2P) as
`Kruskal-Wallis analysis ofvariance (ANOVA) with Dunn’s multiple comparisons
`a transmembrane-anchored protein with the native furin cleavage
`test. Inb, for weightloss, all comparisonsare with PBS control mice at the same
`site (mRNA-1273)was initiatedin parallel with preclinical evaluation.
`time point by two-sided Mann-WhitneyU-test. **P< 0.01,****P< 0.0001. Data
`This led to a first-in-human phaseIclinical trial starting on 16 March
`are GMT+geometrics.d.(a,c) ormean+s.d.(b, d). Inc, thedottedline
`2020, 66 days after the viral sequence wasreleased, and a phase lltrial
`representsassaylimit ofdetection.
`74 days later on 29 May 2020 (Extended DataFig. 2). Expression and
`antigenicity of the S(2P) antigen delivered by mRNAwasconfirmed
`in vitro before vaccinationofthe first human participant (Extended
`Data Fig. 3), and immunogenicity of mMRNA-1273 was documentedin
`several mousestrains. The results of those studies are detailed here.
`
` Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 3 of 22 PagelD #: 12580
`
`Fig. 2| mRNA-1273 elicits robust binding and
`pseudovirus-neutralizing antibody responsesin
`multiple mousestrains. a-f, BALB/c) (a,d), CS7BL/6)
`(b, e) or B6C3F1/J (c, f) mice (nr =10 per group) were
`ad
`5. 2
`5_
`
`
`
`5 £ immunized at weeksO and3with 0.01 (green), 0.13 £ 6 5 £ 6
`g 5
`8 5 5
`i 5 5
`(blue)or1pg(red)mRNA-1273.Control BALB/c] mice
`3 3
`8 3 4
`8 z 4
`wereadministered PBS(grey).Serawerecollected
`5
`5
`3
`5
`3
`2weekspost-prime(unfilled circles) and2weeks
`=
`=
`24
`=
`QM
`post-boost(filled circles) and assessed for SARS-CoV-2
`S-specific|gG byenzyme-linked immunosorbentassay
`(ELISA)(a-c),and forpost-boostsera, neutralizing
`antibodies against homotypicSARS-CoV-2
`pseudovirus(d-f). In a—c,timepointswerecompared
`signed-ranktest, and doses were compared post-boost
`by Kruskal-Wallis ANOVA with Dunn’s multiple
`comparisonstest.In d-f, Vaccine groups were
`compared by two-sided Mann-Whitney U-test.
`*P< 0.05, **P< 0.01,***P< 0.001, ****P< 0.0001. Data
`are presented as GMT+ geometric s.d. Dotted lines
`represent assay limits ofdetection.
`
`2
`
`1
`
`PBS 0.1 1
`mRNA-1273
`dose (ug)
`
`2
`
`1
`
`4
`o1
`mRNA-1273
`dose (ug)
`
`568 | Nature | Vol586 | 22 October 2020
`
`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 3 of 22 PageID #: 12580
`Article
`
`
`
`4
`PBS 0.01 0.1
`MERS S(2P)
`mRNAdose (ug)
`
`S&
`
`#
`
`Lung viral load
`”
`
`d
`
`Lung haemorrhage
`
`Jo PBS 0.01_°0.1
`
`g 4
`He
`i
`a
`o'
`
`=
`
`Aad
`aa
`
`PBS 0.010.111
`MERS S(2P)
`MRNA dose (ug)
`
`1
`MERS S(2P)
`mRNAdose (ug)
`
`c
`
`g
`—
`;
`
`a3
`
`preparednessefforts, we have studied MERS-CoVas a prototype
`Betacoronavirus pathogento optimize vaccine design, dissect the
`humoral immuneresponseto vaccination, and identify mechanisms
`
`a
`
`d
`
`Binding antibodies
`BALB/c
`
`b
`
`Binding antibodies
`CS7BL/6
`
`c
`
`os
`Pes!
`
`Lt,
`O0101
`eeeAny
`Neutralizing antibodies
`5
`
`dt
`‘
`
`iat
`
`—tp
`oN Oe
`ne
`e Nainaie
`oa
`
`Mitt BP
`0.1
`1
`0.01
`Niel
`Neutralizing antibodies
`
`f
`
`
` within each doselevel by two-sided Wilcoxon
`> log(reciprocalIC,
`titre) &
`
`
`log(reciprocalIC,titre)
`oe
`
`

`

`d 03
`_
`=
`@ °?
`8
`FB
`= 0.1
`
`0 coiolliis
`
`.
`
`Naive
`
`1
`0.1
`001
`MRNA-1273 (ug)
`
`ollaall,
`
`1
`0.1
`S(2P)protein (1g)
`+SAS
`
`
`
`
`
`e
`
`0.3
`
`=
`=
`@ 02
`8
`-
`= 0.1
`
`001
`
`1
`
`oO.
`Dose (ug)
`
`CD4 S2 peptide pool
`
`:
`
`.
`
`IFN-y
`TNF
`IL-2
`IL-4
`IL-5
`
`:
`
`a
`
`A.
`
`50.pha oes,
`
`1
`0.1
`S(2P) protein (ug)
`+ SAS
`
`1
`0.1
`0.01
`MRNA-1273 (ug)
`
`Naive
`
`o+ ali...
`
`g
`
`25
`
`~
`¢ °°
`245
`8
`"i 1.0
`Qa
`C05
`
`CD8S2peptide pool
`
`Naive
`
`1
`0.1
`001
`MRNA-1273 (4g)
`
`01
`~S(2P) protein (19)
`+ SAS
`
`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 4 of 22 PageID #: 12581
`
`a
`
`mRNA-1273
`B6C3
`
`b SARS-CoV-2 S(2P) protein + SAS
`B6C3
`
` (IgG2a + IgG2c\igG1 ratio
`B6C3
`
`8
`g
`5 27
`3 = 6
`£5
`& a 4
`223
`ow? 24
`2
`1
`
`1
`O01
`0.01
`mRNAdose (1g)
`
`E38
`5 27
`Be 6
`5
`5
`2 a 4
`@E 3
`o°2
`3s
`1
`
`1
`01
`0.01
`SARS-CoV-2 S(2P)
`dose (1g)
`
`CD4 S1 peptide pool
`
`
`
`
`
` Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 4 of 22 PagelD #: 12581
`
`f -
`.
`— 2.0
`3
`215
`8
`e 1.0
`a
`205
`
`Naive
`
`CD8S1peptide pool
`
`i
`
`1
`0.1
`1
`0.1
`0.01
`MRNA-1273 (ug) S(2P) protein (ug)
`+ SAS
`
`Fig. 3| Immunizations with mRNA-1273 and S(2P) protein, delivered with
`TLR4agonist,elicit S-specific T,1-biased T cell responses. B6C3F1/J mice
`(n=10 per group) were immunizedat weeks0 and 3 with0O.01, 0.1or1pg of
`mRNA-1273 or SARS-CoV-2S(2P)protein with SAS adjuvant. a—c, Sera were
`collected two weekspost-boostand assessed by ELISA for SARS-CoV-2
`S-specific lgG1, and1gG2a and IgG2c. End-pointtitres (a, b) and end-pointtitre
`ratios oflgG2a plus lgG2c tolgG1(c) werecalculated. Ratios were not
`calculated for mice for which end-pointtitres didnot reach the lowerlimit of
`detection (dotted line; N/A). d-g, Seven weekspost-boost, splenocytes were
`isolated from five mice per groupand restimulated with vehicle or pools of
`
`overlapping peptides from SARS-CoV-2S protein in the presence of aprotein
`transport inhibitor cocktail. After 6h,intracellular cytokine staining was
`performed to quantify CD4* and CD8* Tcell responses. Cytokineexpressionin
`the presenceofvehicle only was considered as background and subtracted
`from the responses measured from the S1 and S2 peptide pools for each
`individual mouse.d, e, PercentageofCD4"Tcellsexpressing IFN-y, TNF,IL-2,
`IL-4 and IL-5 inresponseto the S1 (d) and S2(e) peptide pools. f, g, Percentage of
`CD8*T cells expressing IFN-y, TNF and IL-2inresponsetotheS1(f) and S2(g)
`peptide pools.
`
`Immunogenicity was assessed in six-week-old female BALB/c],
`C57BL/6] and B6C3F1/J mice by two intramuscular immunizations
`with 0.01, 0.1 or 1 pg MRNA-1273, separated by a 3-weekinterval.
`mRNA-1273 induced dose-dependent specific S-binding antibod-
`ies after prime and boostin all mousestrains (Fig. 2a—c). Potent
`pseudovirus-neutralizing activity waselicited by 1 pg MRNA-1273,
`reaching reciprocal half-maximal inhibitory concentration (IC,,)
`geometric mean titres (GMTs) of 819 (BALB/cJ), 89 (CS7BL/6]) and
`1,115 (B6C3F1/J) (Fig. 2d-f). Additionally, mice immunized with 1 pg
`mRNA-1273 had robust neutralizing antibodies against pseudovi-
`rusesthat express S protein with the D614G substitution; SARS-CoV-2
`expressing the D614G variantof the S protein has recently become
`dominant around the world? (ExtendedData Fig.4). To further gauge
`immunogenicity across a wide dose range, BALB/c mice were immu-
`nized with 0.0025-20 pg mMRNA-1273, revealing a strong positive cor-
`relation between dose-dependent mRNA-1273-elicited binding and
`pseudovirus-neutralizing antibody responses (ExtendedDataFig.5).
`BALB/c] mice that received a single dose ofmRNA-1273 were evaluated
`to ascertain theutility of a single-dose vaccine. S-binding antibod-
`ies were induced in mice immunizedwith one 1 pg or 10 pg dose of
`mRNA-1273. The 10 pg dose elicited pseudovirus-neutralizing antibody
`
`activity thatincreased between week 2and week4,reaching 315 recip-
`rocal IC.¢g GMT (Extended Data Fig.6a, b). These datademonstrate that
`mRNAexpressing SARS-CoV-2 S(2P) is a potent immunogenand that
`pseudovirus-neutralizing activity can be elicited with a single dose.
`Vaccine-associated enhancedrespiratory disease (VAERD)has been
`associated with T helper2 cell (T,,2)-biased immune responsesin chil-
`dren immunized with whole-inactivated-virus vaccines against RSV
`and measles virus”. A similar phenomenonhasalso been reported in
`some animal models with whole-inactivated vaccines and other types
`of experimental SARS-CoVvaccines”**°. We therefore evaluated the
`balanceof T,,1 and T,,2 cells in immunized mice. Wefirst compared
`levels of S-specific immunoglobulins, IgG2a and IgG2c, and IgG1—
`which are surrogatesofT,,1 and T,,2 responses, respectively—elicited
`by mRNA-1273 with thoseelicited by immunization with SARS-CoV-2
`S(2P) protein using the TLR4 agonist Sigma Adjuvant System (SAS).
`Both immunogenselicited S-binding antibodies in the lgG2a and
`IgG1 subclasses, indicating a balanced T,,1-T,,2 response(Fig. 3a—c,
`Extended DataFig. 7). The S-specific IgG-subclassprofile following a
`single dose of mRNA-1273 (ExtendedData Fig. 6c) was similar to that
`observed following two doses. By contrast, T,,2-biased responses,
`with lower IgG2a/IgG1 ratios, were observed in mice immunized with
`
`Nature | Vol586 | 22 October 2020 | 569
`
`€
`

`

` Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 5 of 22 PagelD #: 12582
`
`Wealso directly measured cytokine patterns in vaccine-induced mem-
`oryT cells by intracellular cytokine staining seven weeksafter the boost
`injection; MRNA-1273-elicited CD4'T cells re-stimulated with S1 or
`S2 peptide pools exhibited a T,,1-dominant response,particularly at
`higher immunogendoses (Fig. 3d, e). Furthermore, 1 pg mMRNA-1273
`induced arobust CD8'T cell responseto the S1 peptide pool(Fig. 3f, g).
`Together, the IgG subclass and T cell cytokine data demonstrate that
`immunization with MRNA-1273elicits balanced T,,1 and T,2 responses,
`in contrast to the T,,2-biased response seen whenusingS protein with
`alum adjuvant, suggesting that mRNAvaccination avoids T,,2-biased
`immune responses, which have been linked to VAERD.
`Protective immunity was assessed in young adult BALB/c] mice
`challenged with mouse-adapted (MA) SARS-CoV-2. SARS-CoV-2 MA
`contains the substitutions Q498Y/P499T in the receptor-binding
`domain*. The substitutions enable the virus to bind to the mouse
`angiotensin-converting enzyme 2 (ACE2) receptor and infect and
`replicate in the upper and lowerrespiratory tract”. BALB/c] mice that
`received two 1 pg doses of mRNA-1273 were completely protected from
`viral replication inlungs after challenge 5 or 13 weeks after boostinjec-
`tion (Fig.4a, Extended DataFig. 9a). mRNA-1273-induced immunity also
`resulted in undetectable viral replication in nasal turbinates in 6 out of
`7 mice (Fig. 4b, Extended DataFig. 9b). The efficacy of mRNA-1273 was
`dose-dependent; two 0.1 pg doses of mRNA-1273 reduced lungviral
`load by about 100-fold, whereas two 0.01 pg doses reduced lungviral
`load by about 3-fold (Fig. 4a). Of note, mice challenged 7 weeks after
`asingle dose of1or 10 pg MRNA-1273 were alsocompletely protected
`againstlungviral replication (Fig. 4c). Challenging animals immunized
`with subprotective dosesprovides an orthogonal assessmentofsafety
`signals such asincreased clinicalillness or pathology. Similar observa-
`tions with MERS-CoVS(2P) mRNA,mice immunized with subprotective
`0.1 or 0.01 1g doses of MRNA-1273 showed noevidence of enhanced
`lung pathologyor excessive mucus production(Fig.4d). Insummary,
`mRNA-1273is immunogenic,efficacious and does not produce evidence
`ofVAERD whengivenat subprotective doses in mice.
`Here we have shownthat 1 pg of MRNA-1273is sufficient to induce
`robust pseudovirus-neutralizing activity and CD8T cell responses,
`balanced T,,1-T,,2 antibody isotype responses,and protection from
`viral replication for more than three monthsfollowing a prime-
`boost regimen similar to the one being tested in humans.Thelevel of
`pseudovirus-neutralizing activity induced by 14g MRNA-1273in miceis
`similar in magnitudeto that induced by 100 pg mRNA-1273 inhumans*,
`whichis the dose selected for mRNA-1273 to advanceinto phaseIII
`clinical trials. The inclusion oflower subprotective doses demonstrates
`the dose-dependenceofantibody,T,,1 CD4T cell responses andprotec-
`tion, suggesting that immune correlates of protection can be further
`elucidated. Animal studies supporting candidate SARS-CoV-2 vaccines
`throughclinicaltrials aim to demonstrateelicitation of potent protec-
`tive immune responsesas well as to show that subprotective responses
`do not cause VAERD*. Subprotective doses ofmRNA-1273did not prime
`mice for enhancedimmunopathology following challenge. Moreover,
`the induction of protective immunity following a single dose suggests
`single-dose administration of this vaccine could be considered in the
`outbreak setting. These data, combined with immunogenicity data
`from non-humanprimates and human participants of early phase I
`clinical trials, have been used to inform the dose and regimen ofmRNA-
`1273 in advanced clinical efficacytrials.
`The COVID-19 pandemic of2020is the widely predicted ‘pathogen X
`event’?“*, Here we providea paradigmfor rapid vaccine development.
`Combining structure-guided stabilization ofthe MERS-CoVS protein
`with a fast, scalable and safe mRNA-LNPvaccine platform has led toa
`generalizable vaccine solution for Betacoronavirus and acommercial
`mRNAvaccine delivery platform; these developments enabled a rapid
`response to the COVID-19 outbreak. This response demonstrates how
`new technology-driven concepts such as synthetic vaccinology can
`facilitate a vaccine development programmeinitiated on the basis of
`
`g
`:
`5
`8
`
`: :
`ra
`3 0
`
`3.
`
`mRNA-1273
`dose (1g)
`
`2x0.01pgmRNA-1
`
`
`Bee&ae4beet
`
`
`
`
`
`Fig. 4|mRNA-1273 protects mice from upper- and lower-airway SARS-CoV-2
`infection. a,b, BALB/c) mice (n=10 per group) immunized at weeks 0 and3
`with 0.01 pg (green), 0.1 pg (blue) or1 pg (red) mRNA-1273 or PBS were
`challenged with SARS-CoV-2 MAfive weekspost-boost. c, Other groups were
`immunizedwith single dosesof0.1,1g (blue), 1g (red) or 10 pg (purple)
`mRNA-1273 and challenged 7 weeksafter immunization. Two daysafter
`challenge, at peakviral load, mouse lungs(a, c) and nasal turbinates (b) were
`collected from five mice per groupto measureviral titres. a—c, Dataare
`presented as GMT + geometrics.d. and dotted lines represent assay limits of
`detection. Group comparisons were made by Kruskal-Wallis ANOVA with
`Dunn’s multiple comparisonstest. **P< 0.01,***P< 0.001. d, At days 2and4
`after challenge, lung sections from 5 mice pergroup werestained with
`haematoxylin and eosin, and representative photomicrographs(original
`magnification x4 (scale bars, 600 pm) and 10 (scale bars, 300 um)as
`indicated) from each groupwith detectablevirusin lung are shown. Day 2lungs
`from PBS control mice demonstrated moderate-to-severe, predominantly
`neutrophilic inflammation present within and surrounding small bronchioles
`(arrowheads); alveolar capillaries were markedly expandedbyinfiltrating
`inflammatorycells. In the 0.01 pg two-dose group,inflammation was minimal
`to absent. Inthe 0.1,1g two-dose group,occasional areasofinflammation
`intimately associated with small airways (bronchioles) and adjacent
`vasculature (arrowheads) wereseen, primarily composed ofneutrophils. In the
`single-dose 0.1 1g group, there were mild patchy expansionsofalveolar septae
`by mononuclear and polymorphonuclear cells. At day 4, lungs from PBScontrol
`mice exhibited moderate-to-marked expansionofalveolar septae(interstitial
`pattern) with decreased prominenceofadjacentalveolarspaces.Inthe 0.01pg
`two-dose group,inflammation was minimalto absent. Lungsin the 0.1p1g
`two-dose group showedmild, predominantly lymphocytic inflammation,
`associated with bronchioles and adjacentvasculature (arrowheads).In the
`single-dose 0.1 1g group there wasmild, predominantly lymphocytic
`inflammation around bronchovascular bundles (arrowheads).
`
`SARS-CoV-2 S(2P) protein formulated in alum (Extended Data Fig.8a, b).
`Following restimulation with peptide pools (one poolof overlapping
`peptides for each S subunit, S1 and S2) covering the entire S protein,
`splenocytes from mice immunized with mRNA-1273 secreted more
`IFN-y (a prototypic T,,1 cytokine) than IL-4,IL-5 or IL-13 (classical T,,2
`cytokines), whereas restimulation with SARS-CoV-2S(2P) protein with
`alum adjuvant induceda T,,2-biased response (Extended DataFig.8c, d).
`
`570 | Nature | Vol 586 | 22 October 2020
`
` 6:
`
`
`1x01pg2x01ugmRNA-1273 _mRNA-1273
`
`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 5 of 22 PageID #: 12582
`Article

`
`@ TwodosesmRNA-1273
`
`DB
`
`Twodoses mRNA-1273
`
`One dose mRNA-1273
`
`

`

`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 6 of 22 PageID #: 12583
`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 6 of 22 PagelD #: 12583
`
`
`pathogen sequencesalone". This study also providesa proofofconcept
`forthe prototype-pathogen approachto pandemic preparedness and
`responsethatis predicated on identifying generalizable solutionsfor
`medical counter measures within virus families or genera”, Although
`the response to the COVID-19 pandemic has been unprecedented in
`its speed and breadth,we envision further improvementsin rapid
`responsesto suchthreats. There are 24 othervirus families that are
`knownto infect humans, and sustained investigation ofthose potential
`threatswill improve our readiness for future pandemics“.
`
`Onlinecontent
`
`Any methods,additional references, Nature Research reporting sum-
`maries, source data, extended data, supplementary information,
`acknowledgements, peer review information;details of author con-
`tributions and competing interests; and statements of data and code
`availability are available at https://doi.org/10.1038/s41586-020-2622-0.
`
`1,
`
`2.
`
`3.
`
`4,
`
`5.
`6.
`
`7,
`
`9.
`
`Pallesen,J. et al. Immunogenicity and structuresof a rationally designed prefusion
`MERS-CoVspike antigen. Proc. Natl Acad. Sci. USA 114, E7348-E7357 (2017).
`Korber, B. et al. Tracking changes in SARS-CoV-2 spike: evidence that D614G increases
`infectivity of the COVID-19 virus. Cell 182, 812-827.e19 (2020).
`Dong, E., Du, H. & Gardner, L. An interactive web-based dashboard to track COVID-19 in
`real time. Lancetinfect. Dis. 20, 533-534 (2020).
`Keni, R., Alexander, A., Nayak, P. G., Mudgal, J. & Nandakumar, K. COVID-19: emergence,
`spread, possible treatments, and global burden.Front. Public Health 8, 216 (2020).
`Graham, B. S. Rapid COVID-19 vaccine development. Science 368, 945-946 (2020).
`Graham, B.S., Gilman, M.S, A. & McLellan, J. S. Structure based vaccine antigen design.
`Annu. Rev. Med. 70, 91-104 (2019).
`McLellan,J. S. et al. Structure of RSV fusion glycoprotein trimer bound toa
`prefusion-specific neutralizing antibody. Science 340,1113-1117 (2013).
`8. McLellan, J. S. et al. Structure-based design of a fusion glycoprotein vaccine for
`respiratory syncytialvirus. Science 342, 592-598 (2013).
`Crank, M. C. et al. A proof of conceptfor structure based vaccine design targeting RSV in
`humans. Science 365, 505-509 (2019).
`10. Gilman, M.S. A. et al. Rapid profiling of RSV antibody repertoires from the memoryB cells
`of naturally infected adult donors. Sci, immunol.1, eaaj1879 (2016).
`11. Walls, A. C. et al. Cryo-electron microscopystructure of a coronavirus spike glycoprotein
`trimer. Nature 531, 114-117 (2016).
`12. Kirchdoerfer, R. N. et al. Pre-fusion structure of a human coronavirusspike protein. Nature
`531, 118-121 (2016).
`13. Graham,B.S. & Sullivan, N.J. Emerging viral diseases from a vaccinology perspective:
`preparing for the next pandemic.Nat. immunol. 19, 20-28 (2018).
`
`19.
`
`14. Graham, B. S. & Corbett, K. S. Prototype pathogen approachfor pandemic preparedness:
`world onfire. J. Clin. invest. 130, 3348-3349 (2020).
`15. Menachery,V. D. et al. A SARS-like cluster of circulating bat coronaviruses shows
`potential for human emergence.Nat. Med. 21, 1508-1513 (2015).
`16. Menachery,V. D. et al. SARS-like WIV1 CoV poised for human emergence. Proc. Nat! Acad.
`Sci, USA 113, 3048-3053 (2016).
`17. Graham, B.S., Mascola, J. R. & Fauci, A. S. Novel vaccine technologies: essential
`componentsof an adequate response to emergingviral diseases. J. Am. Med. Assoc. 319,
`1431-1432 (2018).
`18. Dowd, K. A. et al. Rapid developmentof a DNA vaccinefor Zika virus. Science 354,
`237-240 (2016).
`Pardi, N., Hogan, M.J., Porter, F. W. & Weissman, D. mRNA vaccines—a newera in
`vaccinology. Nat. Rev. Drug Discov. 17, 261-279 (2018).
`20. Hassett, K. J. et al. Optimization oflipid nanoparticles for intramuscular administration of
`mRNAvaccines.Mol. Ther. Nucleic Acids 15, 1-11 (2019).
`21. Mauger, D. M. et al. mRNAstructure regulates protein expression through changesin
`functional half-life. Proc. Natl Acad. Sci. USA 116, 24075-24083(2019).
`22. Cockrell, A. S. et al. A mouse modelfor MERS coronavirus-induced acute respiratory
`distress syndrome.Nat. Microbiol. 2, 16226 (2016).
`23. Wrapp,D. et al. Cryo-EM structure of the 2019-nCoVspikein the prefusion conformation.
`Science 367, 1260-1263 (2020).
`24, Freeman, B.etal. Validation of a SARS-CoV-2 spike protein ELISA for use in contact investigations
`and serosurveillance. Preprint at https://doi.org/10.1101/2020.04.24.057323 (2020).
`25. Klumpp-Thomas,C.et al. Standardization of enzyme-linked immunosorbentassaysfor
`serosurveysof the SARS CoV-2 pandemicusingclinical and at home blood sampling.
`Preprintat https://doi.org/10.1101/2020,05.21.20109280 (2020).
`26. Kim, H.W.et al. Respiratory syncytial virus disease in infants despite prior administration
`of antigenic inactivated vaccine. Am.J. Epidemiol. 89, 422-434 (1969).
`Fulginiti, V. A., Eller, J. J., Downie, A. W, & Kempe, C. H. Altered reactivity to measles virus.
`Atypical measlesin children previously immunized with inactivated measles virus
`vaccines.J, Am. Med, Assoc. 202, 1075-1080(1967).
`28. Bolles, M. et al. A double inactivated severe acute respiratory syndrome coronavirus
`vaccine provides incomplete protection in mice and induces increased eosinophilic
`proinflammatory pulmonary response upon challenge.J. Virol. 85, 12201-12215 (2011).
`29, Czub, M., Weingartl, H., Czub, S., He, R. & Cao,J. Evaluation of modified vaccinia virus
`Ankara based recombinant SARSvaccinein ferrets. Vaccine 23, 2273-2279 (2005).
`30. Deming,D. et al. Vaccine efficacy in senescent mice challenged with recombinant
`SARS-CoVbearing epidemic and zoonotic spike variants. PLoS Med 3, E525 (2006).
`31. Hou, Y.J. et al. SARS-CoV-2 reverse genetics reveals a variable infection gradientin the
`respiratory tract. Cell 182, 429-446 (2020).
`32. Dinnon,K. H. et al. A mouse-adapted modelof SARS CoV-2 to test COVID-19
`countermeasures. Nature https://doi.org/10.1038/s41586-020-2708-8 (2020).
`Jackson,L. A. et al. An mRNAvaccine against SARS CoV-2—preliminary report. N. Engl. J.
`Med. Moa2022483 (2020).
`
`27.
`
`33.
`
`Publisher's note Springer Nature remains neutralwith regard to jurisdictional claims in
`published mapsandinstitutional affiliations.
`
`© This is a U.S. government work and not under copyright protectionin the U.S.; foreign
`copyright protection may apply 2020
`
`Nature | Vol586 | 22 October 2020 | 571
`
`

`

`Case 1:22-cv-00252-MSG Document 193-5 Filed 01/16/24 Page 7 of 22 PageID #: 12584
`Article
`
`with 10% FBS, 2mM glutamine and 1% penicillin-streptomycin at 37 °C
`and 5% CO,. Vero E6cells used in plaque assays to determine lung and
`nasal turbinate viraltitres were cultured in DMEM supplemented with
`10% Fetal Clone Il and 1% antibiotic-antimycotic at 37 °C and 5% CO2.
`Vero E6 cells used in plaque-reduction neutralization test (PRNT)
`assays were cultured in DMEM supplemented with 10% Fetal CloneII
`and amphotericin B (0.25 pg mI“) at 37 °C and 5% CO,. Lentivirus encod-
`ing hACE2-P2A-TMPRSS2 was madeto generate A549-hACE2-TMPRSS2
`cells, which were maintained in DMEM supplemented with 10% FBS
`and 1 pg mI“ puromycin. Expi293 cells were maintained in the manu-
`facturer’s suggested medium. BHK-21/WI-2 cells were obtained from
`Kerafast and cultured in DMEM with 5% FBS at 37 °C and 6-8% CO.,Cell
`lines were not authenticated.All cells lines were tested for mycoplasma
`and remained negative.
`
`In vitro mRNAexpression
`HEK293Tcells were transiently transfected with mRNA encoding
`SARS-CoV-2 wild-type S or S(2P) protein using a TranIT mRNAtrans-
`fection kit (Mirus). After 24 h, the cells were collected and resuspended
`in fluorescence-activated cell sorting (FACS) buffer (1x PBS, 3% FBS,
`0.05% sodium azide). To detect surface-protein expression,the cells
`were stained with 10 pg ml" ACE2-Flag (Sigma) or 10 pg ml’ CR3022
`in FACS bufferfor 30 min onice. Thereaft

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