Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
METHOD OF PROVIDING SAFE ADMINISTRATION OF ADENOVIRAL VECTORS ENCODING A ZIKA VIRUS ANTIGEN
Document Type and Number:
WIPO Patent Application WO/2019/099970
Kind Code:
A1
Abstract:
Provided are methods for generating an immune response against a Zika virus in a human subject in need thereof. The methods comprise administering to the subject a pharmaceutical composition comprising adenoviral vectors encoding a Zika virus antigen and a pharmaceutically acceptable carrier.

Inventors:
BAROUCH DAN (US)
DOUOGUIH MACAYA (US)
PICCHIO GASTON (US)
Application Number:
PCT/US2018/061743
Publication Date:
May 23, 2019
Filing Date:
November 19, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JANSSEN PHARMACEUTICALS INC (US)
BETH ISRAEL DEACONESS MEDICAL CT INC (US)
International Classes:
A61K39/12; A61P31/14
Domestic Patent References:
WO2007104792A22007-09-20
WO2017214596A12017-12-14
WO2006040330A22006-04-20
WO2007104792A22007-09-20
WO2000070071A12000-11-23
WO2005071093A22005-08-04
WO2010086189A22010-08-05
WO2010085984A12010-08-05
WO2003104467A12003-12-18
WO2004001032A22003-12-31
WO2012082918A12012-06-21
Foreign References:
US20170036900W2017-06-09
US7270811B22007-09-18
US6083716A2000-07-04
Other References:
P. ABBINK ET AL: "Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys", SCIENCE, vol. 353, no. 6304, 9 September 2016 (2016-09-09), US, pages 1129 - 1132, XP055314484, ISSN: 0036-8075, DOI: 10.1126/science.aah6157
EUN KIM ET AL: "Preventative Vaccines for Zika Virus Outbreak: Preliminary Evaluation", EBIOMEDICINE, vol. 13, 1 November 2016 (2016-11-01), pages 315 - 320, XP055443543, ISSN: 2352-3964, DOI: 10.1016/j.ebiom.2016.09.028
LUISA BARZON ET AL: "Current views on Zika virus vaccine development", EXPERT OPINION ON BIOLOGICAL THERAPY, vol. 17, no. 10, 26 June 2017 (2017-06-26), ASHLEY, LONDON; GB, pages 1185 - 1192, XP055550338, ISSN: 1471-2598, DOI: 10.1080/14712598.2017.1346081
ABBINK PETER ET AL: "Comparative seroprevalence and immunogenicity of six rare serotype recombinant adenovirus vaccine vectors from subgroups B and D", JOURNAL OF VIROLOGY, THE AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 81, no. 9, 1 May 2007 (2007-05-01), pages 4654 - 4663, XP002451979, ISSN: 0022-538X
L. R. BADEN ET AL: "First-in-Human Evaluation of the Safety and Immunogenicity of a Recombinant Adenovirus Serotype 26 HIV-1 Env Vaccine (IPCAVD 001)", THE JOURNAL OF INFECTIOUS DISEASES, vol. 207, no. 2, 2 November 2012 (2012-11-02), pages 240 - 247, XP055190590, ISSN: 0022-1899, DOI: 10.1093/infdis/jis670
FREEK COX ET AL: "Adenoviral vector type 26 encoding Zika virus (ZIKV) M-Env antigen induces humoral and cellular immune responses and protects mice and nonhuman primates against ZIKV challenge", PLOS ONE, vol. 13, no. 8, 24 August 2018 (2018-08-24), pages e0202820, XP055551627, DOI: 10.1371/journal.pone.0202820
ABBINK PETER ET AL: "Zika virus vaccines", NATURE REVIEWS. MICROBIOLOGY, NATURE PUBLISHING GROUP, GB, vol. 16, no. 10, 19 June 2018 (2018-06-19), pages 594 - 600, XP036624828, ISSN: 1740-1526, [retrieved on 20180619], DOI: 10.1038/S41579-018-0039-7
PETER ABBINK ET AL: "Durability and correlates of vaccine protection against Zika virus in rhesus monkeys", SCIENCE TRANSLATIONAL MEDICINE, vol. 9, no. 420, 13 December 2017 (2017-12-13), US, pages eaao4163, XP055550327, ISSN: 1946-6234, DOI: 10.1126/scitranslmed.aao4163
CÉSAR LÓPEZ-CAMACHO ET AL: "Rational Zika vaccine design via the modulation of antigen membrane anchors in chimpanzee adenoviral vectors", NATURE COMMUNICATIONS, vol. 9, no. 1, 22 June 2018 (2018-06-22), XP055551656, DOI: 10.1038/s41467-018-04859-5
CLINICALTRIALS.GOV: "NCT03356561: A Study to Evaluate the Safety, Reactogenicity and Immunogenicity of Ad26.ZIKV.001 in Healthy Adult Volunteers", 29 November 2017 (2017-11-29), XP055552132, Retrieved from the Internet [retrieved on 20190205]
SMITH; WATERMAN, ADV. APPL. MATH., vol. 2, 1981, pages 482
NEEDLEMAN; WUNSCH, J. MOL. BIOL., vol. 48, 1970, pages 443
PEARSON; LIPMAN, PROC. NAT'L. ACAD. SCI. USA, vol. 85, 1988, pages 2444
"Wisconsin Genetics Software Package", GENETICS COMPUTER GROUP
"Current Protocols in Molecular Biology", CURRENT PROTOCOLS
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
HENIKOFF; HENIKOFF, PROC. NATL. ACAD. SCI. USA, vol. 89, 1989, pages 10915
KARLIN; ALTSCHUL, PROC. NAT'L. ACAD. SCI. USA, vol. 90, 1993, pages 5873 - 5787
ABBINK ET AL., VIROL, vol. 81, no. 9, 2007, pages 4654 - 63
"GenBank", Database accession no. EF 153474
VOGELS ET AL., J VIROL, vol. 77, no. 15, 2003, pages 8263 - 71
"GenBank", Database accession no. AC 000019
FARINA ET AL., J VIROL, vol. 75, 2001, pages 11603 - 13
COHEN ET AL., J GEN VIROL, vol. 83, 2002, pages 151 - 55
KOBINGER ET AL., VIROLOGY, vol. 346, 2006, pages 394 - 401
TATSIS ET AL., MOLECULAR THERAPY, vol. 15, 2007, pages 608 - 17
BANGARI; MITTAL, VACCINE, vol. 24, 2006, pages 849 - 62
LASARO; ERTL, MOL THER, vol. 17, 2009, pages 1333 - 39
HAVENGA ET AL., J GEN VIROL, vol. 87, 2006, pages 2135 - 43
"GenBank", Database accession no. AC _000019
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"Encyclopedia of Pharmaceutical Technology", 2006, INFORMA HEALTHCARE
"Remington: The Science and Practice of Pharmacy", 2005
MODJARRAD ET AL., THE LANCET, vol. 391, no. 10120, 10 February 2018 (2018-02-10), pages 563 - 571
ABBINK ET AL., SCIENCE, vol. 353, no. 6304, 9 September 2016 (2016-09-09), pages 1129 - 1132
ABBINK ET AL., SCI TRANSL MED., vol. 9, no. 420, 13 December 2017 (2017-12-13)
Attorney, Agent or Firm:
BARANIAK, Andrew, P. et al. (US)
Download PDF:
Claims:
CLAIMS

It is claimed:

1. A method for generating an immune response against a Zika virus in a human subject in need thereof, the method comprising administering to the subject a

pharmaceutical composition comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier, wherein about 5 x 1010 adenoviral vectors to about 1 x 1011 adenoviral vectors are administered per dose to the subject in need thereof.

2. The method of claim 1, wherein about 5 x 1010 adenoviral vectors are

administered per dose to the subject in need thereof.

3. The method of claim 1, wherein about 1 x 1011 adenoviral vectors are

administered per dose to the subject in need thereof.

4. The method of any one of claims 1-3, wherein the pharmaceutical composition is administered via an intramuscular injection to the subject in need thereof.

5. The method of any one of claims 1-4, wherein the pharmaceutical composition is administered to the human subject as a single dose.

6. The method of any one of claims 1-4, wherein the pharmaceutical composition is administered to the human subject as a double dose.

7. The method of claim 6, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart.

8. The method of claim 7, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about eight weeks apart.

9. The method of any one of claims 1-8, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2, 4, 6, 8, 10, or 12.

10. The method of claim 9, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2.

11. The method of any one of claims 1-10, wherein the adenoviral vectors are of the Ad26 serotype.

Description:
METHOD OF PROVIDING SAFE ADMINISTRATION OF ADENOVIRAL VECTORS ENCODING A ZIKA VIRUS ANTIGEN

FIELD OF THE INVENTION

[0001] This invention relates to biotechnology. More particularly, to methods of administering adenovirus viral particles comprising an optimized, non-naturally occurring Zika virus (ZIKV) nucleic acid molecule to a subject in need thereof for preventing a ZIKV infection and/or symptoms caused by a ZIKV.

REFERENCE TO SEQUENCE LISTING SUBMITTED ELECTRONICALLY

[0002] This application contains a sequence listing, which is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file name“688097.564WO Sequence Listing” and a creation date of November 14, 2018, and having a size of 186 kb. The sequence listing submitted via EFS-Web is part of the specification and is herein incorporated by reference in its entirety.

BACKGROUND OF THE INVENTION

[0003] Zika virus (ZIKV) is a flavivirus that is responsible for an unprecedented current epidemic in Brazil and the Americas. ZIKV has been causally associated with microcephaly, intrauterine growth restriction, and other birth defects in humans and in murine models. ZIKV is believed to cause neuropathology in developing fetuses by crossing the placenta and targeting cortical neural progenitor cells, leading to impaired neurogenesis and resulting in microcephaly and other congenital malformations.

[0004] The World Health Organization declared the clusters of microcephaly and neurological disorders and their association with ZIKV infection to be a global public health emergency on February 1, 2016. ZIKV also has been associated with neurologic conditions such as Guillain-Barre syndrome. While the rapid development of a safe and effective ZIKV vaccine is a global health priority, very little is currently known about ZIKV immunology and mechanisms of immune protection.

[0005] Accordingly, there is an unmet need in the field for ZIKV vaccines. BRIEF SUMMARY OF THE INVENTION

[0006] Provided herein are methods for generating an immune response against a Zika virus in a human subject in need thereof. A method according to an embodiment of the invention comprises administering to the subject a pharmaceutical composition comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier, wherein about 1 x 10 10 adenoviral vectors to about 5 x 10 11 adenoviral vectors, preferably about 5 x 10 10 adenoviral vectors to about 1 x 10 11 adenoviral vectors are administered to the subject in need thereof. In a preferred embodiment, about 5 x 10 10 adenoviral vectors are administered per dose to the human subject in need thereof. In another preferred embodiment, about 1 x 10 11 adenoviral vectors are administered per dose to the human subject in need thereof.

[0007] In certain embodiments, the pharmaceutical composition is administered via an intramuscular injection to the human subject in need thereof. In certain embodiments, the pharmaceutical composition is administered to the human subject as a single dose. In certain embodiments, the pharmaceutical composition is administered to the human subject as a double dose. The first and second dose of the pharmaceutical composition can be administered to the human subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart. In certain embodiments, the first and second dose of the pharmaceutical composition can be administered to the human subject about eight weeks apart.

[0008] In certain embodiments, the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2, 4, 6, 8, 10, or 12. In certain embodiments, the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2.

[0009] In certain embodiments, the adenoviral vectors are of the Ad26 serotype.

BRIEF DESCRIPTION OF THE DRAWINGS

[0010] The foregoing summary, as well as the following detailed description of the invention, will be better understood when read in conjunction with the appended drawings. It should be understood that the invention is not limited to the precise embodiments shown in the drawings. [0011] FIGS 1 A and 1B show the ZIKV neutralizing antibody response (assessed by MN50 VNA) in serum from clinical study participants at the indicated study timepoints (days 15 and 29 on FIG. 1A and days 57, 71 and 85 on FIG. 1B) for each of the tested regimens. Doses were given at days 1 and 57. Responders are depicted on FIGS. 1 A and 1B with filled circles, non-responders are depicted with open light grey circles. Pooled baseline is depicted with dark grey circles on FIG. 1 A on the left-hand side of the graph. LD stands for Low Dose which stands for a dose of 5xl0 10 viral particles (vp) of Ad26.ZIKV.00l; HD stands for High Dose, which stands for a dose of lxlO 11 vp Ad26.ZIKV.00l, PL stands for Placebo.

DETAILED DESCRIPTION OF THE INVENTION

[0012] Various publications, articles and patents are cited or described in the background and throughout the specification; each of these references is herein incorporated by reference in its entirety. Discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is for the purpose of providing context for the invention. Such discussion is not an admission that any or all of these matters form part of the prior art with respect to any inventions disclosed or claimed.

[0013] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention pertains. Otherwise, certain terms used herein have the meanings as set forth in the specification.

[0014] It must be noted that as used herein and in the appended claims, the singular forms“a,”“an,” and“the” include plural reference unless the context clearly dictates otherwise.

[0015] Unless otherwise stated, any numerical values, such as a concentration or a concentration range described herein, are to be understood as being modified in all instances by the term“about.” Thus, a numerical value typically includes ± 10% of the recited value. For example, a concentration of 1 mg/mL includes 0.9 mg/mL to 1.1 mg/mL. Likewise, a concentration range of 1% to 10% (w/v) includes 0.9% (w/v) to 11% (w/v). As used herein, the use of a numerical range expressly includes all possible subranges, all individual numerical values within that range, including integers within such ranges and fractions of the values unless the context clearly indicates otherwise.

[0016] Unless otherwise indicated, the term“at least” preceding a series of elements is to be understood to refer to every element in the series. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the invention.

[0017] It should also be understood that the terms“about,”“approximately,” “generally,”“substantially” and like terms, used herein when referring to a dimension or characteristic of a component of the preferred invention, indicate that the described dimension/characteristic is not a strict boundary or parameter and does not exclude minor variations therefrom that are functionally the same or similar, as would be understood by one having ordinary skill in the art. At a minimum, such references that include a numerical parameter would include variations that, using mathematical and industrial principles accepted in the art (e.g., rounding, measurement or other systematic errors, manufacturing tolerances, etc.), would not vary the least significant digit.

[0018] As used herein, the terms“comprises,”“comprising,”“includes,”“incl uding,” “has,”“having,”“contains” or“containing,” or any other variation thereof, will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers and are intended to be non-exclusive or open-ended. For example, a composition, a mixture, a process, a method, an article, or an apparatus that comprises a list of elements is not necessarily limited to only those elements but can include other elements not expressly listed or inherent to such composition, mixture, process, method, article, or apparatus. Further, unless expressly stated to the contrary,“or” refers to an inclusive or and not to an exclusive or. For example, a condition A or B is satisfied by any one of the following: A is true (or present) and B is false (or not present), A is false (or not present) and B is true (or present), and both A and B are true (or present).

[0019] As used herein, the conjunctive term“and/or” between multiple recited elements is understood as encompassing both individual and combined options. For instance, where two elements are conjoined by“and/or,” a first option refers to the applicability of the first element without the second. A second option refers to the applicability of the second element without the first. A third option refers to the applicability of the first and second elements together. Any one of these options is understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or” as used herein.

Concurrent applicability of more than one of the options is also understood to fall within the meaning, and therefore satisfy the requirement of the term“and/or.”

[0020] As used herein, the term“consists of,” or variations such as“consist of’ or “consisting of,” as used throughout the specification and claims, indicate the inclusion of any recited integer or group of integers, but that no additional integer or group of integers can be added to the specified method, structure, or composition.

[0021] As used herein, the term“consists essentially of,” or variations such as“consist essentially of’ or“consisting essentially of,” as used throughout the specification and claims, indicate the inclusion of any recited integer or group of integers, and the optional inclusion of any recited integer or group of integers that do not materially change the basic or novel properties of the specified method, structure or composition. See M.P.E.P. § 2111.03.

[0022] As used herein,“subject” or“patient” means any animal, preferably a mammal, most preferably a human, to whom will be or has been administered a vaccine by a method according to an embodiment of the invention. The term“mammal” as used herein, encompasses any mammal. Examples of mammals include, but are not limited to, cows, horses, sheep, pigs, cats, dogs, mice, rats, rabbits, guinea pigs, monkeys, humans, etc., more preferably a human.

[0023] As used herein,“a method of providing safe administration” means a method of administration that is effective in generating an immune response against a Zika virus without causing unacceptable adverse events, when administered to a subject.

[0024] As used herein, the phrases“unacceptable adverse events” and“unacceptable adverse reaction” shall mean all harm or undesired outcomes associated with or caused by administration of a pharmaceutical composition or therapeutic, and the harm or undesired outcome reaches such a level of severity that a regulatory agency deems the pharmaceutical composition or therapeutic unacceptable for the proposed use. Examples of unacceptable adverse events or reactions when used in the context of administration of adenoviral particles comprising a nucleic acid molecule encoding a Zika virus antigen can include, but is not limited to, swelling, injection side pain, headache, malaise, muscle ache, nausea, and fever.

[0025] In certain embodiments,“safe treatment” and“safe administration” when used with respect to administration of adenoviral vectors comprising a nucleic acid molecule encoding a Zika virus antigen means reduced adverse events including, but not limited to, swelling, injection side pain, headache, malaise, muscle ache, nausea, and fever.

[0026] The terms“identical” or percent“identity,” in the context of two or more nucleic acids or polypeptide sequences (e.g., Zika antigen polypeptides and

polynucleotides that encode them), refer to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same, when compared and aligned for maximum correspondence, as measured using one of the following sequence comparison algorithms or by visual inspection.

[0027] For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are input into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.

The sequence comparison algorithm then calculates the percent sequence identity for the test sequence(s) relative to the reference sequence, based on the designated program parameters.

[0028] Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), by the search for similarity method of Pearson & Lipman, Proc. NatT. Acad. Sci. USA 85:2444 (1988), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, WI), or by visual inspection (see generally, Current Protocols in Molecular Biology, F.M. Ausubel et ak, eds., Current Protocols, a joint venture between Greene Publishing Associates, Inc. and John Wiley & Sons, Inc., (1995 Supplement) (Ausubel)). [0029] Examples of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1990) J. Mol. Biol. 215: 403-410 and Altschul et al. (1997) Nucleic Acids Res. 25: 3389-3402, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. This algorithm involves first identifying high scoring sequence pairs (HSPs) by identifying short words of length W in the query sequence, which either match or satisfy some positive-valued threshold score T when aligned with a word of the same length in a database sequence. T is referred to as the neighborhood word score threshold (Altschul et al, supra). These initial neighborhood word hits act as seeds for initiating searches to find longer HSPs containing them. The word hits are then extended in both directions along each sequence for as far as the cumulative alignment score can be increased.

[0030] Cumulative scores are calculated using, for nucleotide sequences, the

parameters M (reward score for a pair of matching residues; always > 0) and N (penalty score for mismatching residues; always < 0). For amino acid sequences, a scoring matrix is used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide

sequences) uses as defaults a wordlength (W) of 11, an expectation (E) of 10, M=5, N=- 4, and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength (W) of 3, an expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff & Henikoff, Proc. Natl. Acad. Sci. USA 89: 10915 (1989)).

[0031] In addition to calculating percent sequence identity, the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin & Altschul, Proc. NatT. Acad. Sci. USA 90:5873-5787 (1993)). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.1, more preferably less than about 0.01, and most preferably less than about 0.001.

[0032] A further indication that two nucleic acid sequences or polypeptides are substantially identical is that the polypeptide encoded by the first nucleic acid is immunologically cross reactive with the polypeptide encoded by the second nucleic acid, as described below. Thus, a polypeptide is typically substantially identical to a second polypeptide, for example, where the two peptides differ only by conservative

substitutions. Another indication that two nucleic acid sequences are substantially identical is that the two molecules hybridize to each other under stringent conditions.

[0033] As used herein, the term“immune response” or“protective immune response” means that the vaccinated subject is able to control an infection (e.g., a Zika virus (ZIKV) infection) with the pathogenic agent against which the vaccination was done (e.g., a ZIKV antigen). The pathogenic agent can, for example, be an antigenic gene product or antigenic protein, or a fragment thereof. Usually, the subject having developed an“immune response” or a“protective immune response” develops only mild to moderate clinical symptoms or no symptoms at all. Usually, a subject in which an “immune response” or“protective immune response” against a Zika virus has been generated, will not develop disease manifestations or those disease manifestations will be milder, and ultimately the subject will not die as a result of the infection with said virus. In addition, a subject in which an“immune response” or“protective immune response” against a Zika virus has been generated, will have a reduced chance of brain

abnormalities in her infants exposed to the virus in utero.

[0034] By“generating an immune response” or“promoting an immune response” or “provoking an immune response” is meant eliciting a humoral response (e.g., the production of antibodies) or a cellular response (e.g., the activation of T cells, macrophages, neutrophils, and/or natural killer cells) directed against, for example, one or more infective agents (e.g., a virus (e.g., a ZIKV)) or protein targets in a subject to which the pharmaceutical composition (e.g., an immunogenic composition or vaccine) has been administered. [0035] By“immunogen” or“antigen” is meant any polypeptide that can induce an immune response in a subject upon administration. In some embodiments, the immunogen or antigen is encoded by a nucleic acid molecule that may be incorporated into, for example, a polynucleotide or vector of the invention, for subsequent expression of the immunogen or antigen (e.g., a gene product of interest, or fragment thereof (e.g., a polypeptide)). In some embodiments, the immunogen is derived from a ZIKV (e.g., a ZIKV from the Asian and /or African lineage (e.g., ZIKV strain BeH815744 (accession number KU365780 (SEQ ID NOs: 15-16))). In some embodiments, the immunogen is administered in the context of a nucleic acid molecule expressing a polypeptide that is derived from a ZIKV (e.g., a ZIKV from the Asian and /or African lineage (e.g., ZIKV strain BeH8l5744 (accession number KU365780 (SEQ ID NOs: 15-16))).

[0036] The term“immunogenic composition” as used herein, is defined as material used to generate an immune response and may confer immunity after administration of the immunogenic composition to a subject.

[0037] By“isolated” is meant separated, recovered, or purified from a component of its natural environment. For example, a nucleic acid molecule or polypeptide of the invention may be isolated from a component of its natural environment by 1% (2%, 3%, 4%, 5%, 6%, 7%, 8% 9% 10%, 20%, 30%, 40%, 50%, 60% 70%, 80%, or 90%) or more.

Nucleic Acid Molecules, Polypeptides, and Vectors of the Invention

[0038] In PCT/US2017/36900, filed on June 9, 2017, entitled“Compositions and Methods for Preventing and Treating Zika Virus Infection,” which is incorporated by reference in its entirety, disclosed are Zika virus (ZIKV) polypeptides that can be used to elicit protective immune responses against a ZIKV infection when administered to a subject (e.g., a mouse or monkey) infected with or likely to be exposed to a ZIKV infection. The ZIKV polypeptides for use in pharmaceutical compositions prepared for administration can include a M-Env, prM-Env, prM-Env.dTM, prM-Env.dStem, Env, Env.dTM, and/or Env.dStem or a portion thereof. Alternatively, the ZIKV polypeptides can be encoded for by a nucleic acid molecule comprised within a vector (e.g., an adenoviral vector). [0039] Table 1 provides the ZIKV derived polypeptide and polynucleotide molecules that can be used in pharmaceutical compositions of the invention for the safe

administration and prevention of a Zika virus infection.

Table 1: ZIKV derived polynucleotide and polypeptide molecules

[0040] The nucleic acid molecules of the invention (Table 1) were designed based on the Zika virus (ZIKV) strain BeH815744 (accession number KU365780 (SEQ ID NOs: 15-16)). The nucleic acid molecules of the invention encode regions of the Zika virus (ZIKV) polyprotein, for example, the membrane and envelope (M-Env) proteins, the pre- membrane and envelope (prM-Env) region, the Env protein alone, or deletion mutants of the prM-Env or Env regions in which the transmembrane (TM) or Stem region have been removed. The nucleic acid molecules of the invention M-Env (SEQ ID NO: 1), prM- Env.dTM (SEQ ID NO: 3), prM-Env.dStem (SEQ ID NO: 5), Env (SEQ ID NO: 7), Env.dTM (SEQ ID NO: 9), and Env.dStem (SEQ ID NO: 11) have been optimized relative to the wild-type BeH815744 nucleotide sequences for improved expression in host cells (e.g., mammalian (e.g., human) host cells) and particle formation, and encode the polypeptides set forth in SEQ ID NOs: 2, 4, 6, 8, 10, or 12, respectively (Table 1). Optimization can include the addition of a leader sequence, such as a Japanese encephalitis virus (JEV) leader sequence (e.g., SEQ ID NO: 13), restriction site (e.g., SEQ ID NOs: 21-22), and/or a Kozak sequence (e.g., SEQ ID NO: 23).

[0041] The prM-Env (full length) (e.g., SEQ ID NOs: 24-25) contains the full-length sequence of the prM-Env region, while prM-Env with JEV Stem/TM (e.g., SEQ ID NOs: 26-27) includes the ZIKV prM signal region of Japanese encephalitis virus (JEV) with the final 98 amino acids comprising the stem and transmembrane regions exchanged with corresponding JEV sequences.

[0042] The nucleic acid molecules have a nucleotide sequence with at least 85% (e.g., at least 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) sequence identity to, all or a portion of any one of SEQ ID NOs: 1, 3, 5, 7, 9, or 11, or a complementary sequence thereof. Alternatively, an isolated nucleic acid molecule has a nucleotide sequence that encodes a ZIKV polypeptide with at least 85% (e.g., at least 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) sequence identity to an amino acid sequence of any one of SEQ ID NOs:

2, 4, 6, 8, 10, or 12.

[0043] The nucleic acid molecules of the invention may be further optimized, such as by codon optimization, for expression in a targeted mammalian subject (e.g., human).

[0044] The nucleic acid molecules can also be inserted into expression vectors, such as an adenovirus vector and incorporated into compositions of the invention. The terms “adenovirus vector” and“adenoviral vector” and“adenoviral particles” are used interchangeably and refer to a genetically-engineered adenovirus that is designed to insert a polynucleotide of interest (e.g., a polynucleotide encoding a ZIKV antigen of the invention) into a eukaryotic cell, such that the polynucleotide is subsequently expressed. Examples of adenoviruses that can be used as a viral vector of the invention include those having, or derived from, the serotypes Ad2, Ad5, Ad 11, Ad 12, Ad24, Ad26, Ad34,

Ad35, Ad40, Ad48, Ad49, Ad50, Ad52 (e.g., RhAd52), and Pan9 (also known as AdC68); these vectors can be derived from, for example, human, chimpanzee (e.g., ChAdl, ChAd3, ChAd7, ChAd8, ChAd2l, ChAd22, ChAd23, ChAd24, ChAd25, ChAd26, ChAd27.l, ChAd28.l, ChAd29, ChAd30, ChAd3 l.l, ChAd32, ChAd33, ChAd34, ChAd35.l, ChAd36, ChAd37.2, ChAd39, ChAd40.l, ChAd4l.l, ChAd42.l, ChAd43, ChAd44, ChAd45, ChAd46, ChAd48, ChAd49, ChAd49, ChAd50, ChAd67, or SA7P), or rhesus adenoviruses (e.g., rhAd5l, rhAd52, or rhAd53).

[0045] “Nucleic acid molecule” or“polynucleotide,” as used interchangeably herein, refer to polymers of nucleotides of any length, and include DNA and RNA. The nucleotides can be deoxyribonucleotides, ribonucleotides, modified nucleotides or bases, and/or their analogs, or any substrate that can be incorporated into a polymer by DNA or RNA polymerase, or by a synthetic reaction. A polynucleotide can comprise modified nucleotides, such as methylated nucleotides and their analogs. If present, modification to the nucleotide structure can be imparted before or after assembly of the polymer. The sequence of nucleotides can be interrupted by non-nucleotide components. A

polynucleotide can be further modified after synthesis, such as by conjugation with a label.

[0046] By“heterologous nucleic acid molecule” is meant a nucleotide sequence that can encode proteins derived or obtained from pathogenic organisms, such as viruses, which can be incorporated into a polynucleotide or vector of the invention. Heterologous nucleic acids can also encode synthetic or artificial proteins, such as immunogenic epitopes, constructed to induce immunity. An example of a heterologous nucleic acid molecule is one that encodes one or more immunogenic peptides or polypeptides derived from a Zika virus (ZIKV). The heterologous nucleic acid molecule is one that is not normally associated with the other nucleic acid molecules found in the polynucleotide or vector into which the heterologous nucleic acid molecule is incorporated.

[0047] A“nucleic acid vaccine” or“DNA vaccine” refers to a vaccine that includes a heterologous nucleic acid molecule under the control of a promoter for expression in a subject. The heterologous nucleic acid molecule can be incorporated into an expression vector, such as a plasmid or an adenoviral vector.

[0048] The term“vaccine” as used herein, is defined as material used to provoke an immune response and that confers immunity for a period of time after administration of the vaccine to a subject.

[0049] A“promoter” is a nucleic acid sequence enabling the initiation of the transcription of a gene sequence in a messenger RNA, such transcription being initiated with the binding of an RNA polymerase on or nearby the promoter.

[0050] A nucleic acid is“operably linked” when it is placed into a structural or functional relationship with another nucleic acid sequence. For example, one segment of DNA can be operably linked to another segment of DNA if they are positioned relative to one another on the same contiguous DNA molecule and have a structural or functional relationship, such as a promoter or enhancer that is positioned relative to a coding sequence so as to facilitate transcription of the coding sequence; a ribosome binding site that is positioned relative to a coding sequence so as to facilitate translation; or a pre- sequence or secretory leader that is positioned relative to a coding sequence so as to facilitate expression of a pre-protein (e.g., a pre-protein that participates in the secretion of the encoded polypeptide). In other examples, the operably linked nucleic acid sequences are not contiguous, but are positioned in such a way that they have a functional relationship with each other as nucleic acids or as proteins that are expressed by them. Enhancers, for example, do not have to be contiguous. Linking can be accomplished by ligation at convenient restriction sites or by using synthetic oligonucleotide adaptors or linkers.

[0051] The polypeptides encoded by vectors useful for the invention are ZIKV polypeptides corresponding to, for example, the membrane and envelope (M-Env) proteins, the pre-membrane and envelope (prM-Env) region, the Env protein alone, or deletion mutants of the prM-Env or Env regions in which the transmembrane (TM) or Stem region has been removed. Exemplary polypeptides encoded by vectors useful for the invention include M-Env (SEQ ID NO: 2), prM-Env. dTM (SEQ ID NO: 4), prM- Env.dStem (SEQ ID NO: 6), Env (SEQ ID NO: 8), Env. dTM (SEQ ID NO: 10), and Env.dStem (SEQ ID NO: 12) and variants having at least 85% (e.g., at least 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100%) sequence identity to, all or a portion of any one of SEQ ID NOs: 2, 4, 6, 8, 10, or 12. The polypeptides of the invention may include at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, or 1800 or more continuous or non-continuous amino acids of any one of SEQ ID NOs: 2, 4, 6, 8, 10, or 12.

[0052] Polypeptides encoded by vectors useful for the invention can also include a signal/leader sequence, such as a Japanese encephalitis virus (JEV) signal/leader sequence (SEQ ID NO: 14). The polypeptides can also be isolated from other components (e.g., components with which the polypeptides are natively associated) and incorporated into compositions of the invention.

[0053] By“portion” or“fragment” is meant a part of a whole. A portion can comprise at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% of the entire length of a polynucleotide or polypeptide sequence region. For polynucleotides, for example, a portion can include at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800 or more contiguous nucleotides of a reference polynucleotide molecule. For polypeptides, for example, a portion can include at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, or 600 or more continuous amino acids of a reference polypeptide molecule.

[0054] In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700 or more consecutive nucleotides of the polynucleotide M- Env (SEQ ID NO: 1). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, or more consecutive nucleotides of the

polynucleotide prM-Env.dTM (SEQ ID NO: 3). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, or more consecutive nucleotides of the polynucleotide prM-Env.dStem (SEQ ID NO: 5). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60, 70, 80, 90,

100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, or more consecutive nucleotides of the polynucleotide Env (SEQ ID NO: 7). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60,

70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, or more consecutive nucleotides of the polynucleotide Env.dTM (SEQ ID NO: 9). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60,

70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, or more consecutive nucleotides of the polynucleotide Env.dStem (SEQ ID NO: 11). In some instances, a fragment of a nucleic acid molecule can include at least 5, 6, 7, 8, 9, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, or more consecutive nucleotides of the polynucleotide prM-Env (full length) (SEQ ID NO: 24). In some instances, a fragment of a nucleic acid molecule can include at least 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, or more consecutive nucleotides of the polynucleotide prM-Env with JEV Stem/TM (SEQ ID NO: 26).

[0055] In some instances, a fragment of a polypeptide can include at least 20, 25, 50,

75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, or more consecutive amino acids of polypeptide M-Env (SEQ ID NO: 2). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90,

100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, or more consecutive amino acids of polypeptide prM-Env. dTM (SEQ ID NO: 4). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, or more

consecutive amino acids of polypeptide prM-Env. dStem (SEQ ID NO: 6). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, or more consecutive amino acids of polypeptide Env (SEQ ID NO: 8). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, or more consecutive amino acids of polypeptide Env. dTM (SEQ ID NO: 10). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, or more consecutive amino acids of polypeptide Env. dStem (SEQ ID NO: 12). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75,

90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or more consecutive amino acids of polypeptide prM- Env (full length) (SEQ ID NO: 25). In some instances, a fragment of a polypeptide can include at least 20, 25, 50, 75, 90, 100, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, or more consecutive amino acids of polypeptide prM-Env with JEV Stem/TM (SEQ ID NO: 27).

[0056] In some instances, administration of a fragment of a polynucleotide (e,g., SEQ ID NOs: 1, 3, 5, 7, 9, 11, 24, and/or 26) and/or a polypeptide (e.g., SEQ ID NOs: 2, 4, 6,

8, 10, 12, 25, and/or 27) of the invention to a subject can illicit an immune response in the subject. [0057] The invention also features recombinant vectors including any one or more of the polynucleotides described above. The vectors of the invention can be used to deliver a nucleic acid expressing an immunogen of the invention (e.g., one of more of SEQ ID NOs: 2, 4, 6, 8, 10, or 12 or variants thereof, having at least 85-99% sequence identity thereto, for example at least greater than 90% sequence identity thereto), and include mammalian, viral, and bacterial expression vectors. The mammalian, viral, and bacterial vectors of the invention can be genetically modified to contain one or more nucleic acid sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, or 11 or variants thereof, having at least 85-99% sequence identity thereto, for example at least greater than 90% sequence identity thereto, and complements thereof.

[0058] Promoters and other expression regulation signals can be selected to be compatible with the host cell for which expression is designed. For example, mammalian promoters include the metallothionein promoter, which can be induced in response to heavy metals, such as cadmium, and the b-actin promoter. A viral promoter, which can be obtained from the genome of a virus, such as, for example, polyoma virus, fowlpox virus, adenovirus (A), bovine papilloma virus, avian sarcoma virus, cytomegalovirus (CMV), a retrovirus, hepatitis-B virus, and Simian Virus 40 (SV40), and human papillomavirus (HPV), can also be used. These promoters are well known and readily available in the art.

[0059] A preferred promoter element is the CMV immediate early promoter. In some embodiments, the expression plasmid is pcDNA3.l+ (Invitrogen, CA, USA). In some embodiments, the expression vector is a viral vector, such as a vector derived from adenovirus or poxvirus.

[0060] Viral genomes provide a rich source of vectors that can be used for the efficient delivery of exogenous genes into the genome of a cell (e.g., a eukaryotic or prokaryotic cell). Viral genomes are particularly useful vectors for gene delivery because the polynucleotides contained within such genomes are typically incorporated into the genome of a target cell by generalized or specialized transduction. These processes occur as part of the natural viral replication cycle, and do not require added proteins or reagents in order to induce gene integration. Examples of viral vectors that can be used to deliver a nucleic acid expressing an immunogen of the invention (e.g., one of more of SEQ ID NOs: 2, 4, 6, 8, 10, or 12 or variants thereof having at least 85-99% sequence identity thereto, for example at least greater than 90% sequence identity thereto) include adenovirus (e.g., Ad2, Ad5, Adl l, Adl2, Ad24, Ad26, Ad34, Ad35, Ad40, Ad48, Ad49, Ad50, Ad52 (e.g., RhAd52), and Pan9 (also known as AdC68)). These adenovirus vectors can be derived from, for example, human, chimpanzee (e.g., ChAdl, ChAd3, ChAd7, ChAd8, ChAd2l, ChAd22, ChAd23, ChAd24, ChAd25, ChAd26, ChAd27.l, ChAd28. l, ChAd29, ChAd30, ChAd3 l. l, ChAd32, ChAd33, ChAd34, ChAd35. l, ChAd36, ChAd37.2, ChAd39, ChAd40. l, ChAd4l. l, ChAd42. l, ChAd43, ChAd44, ChAd45, ChAd46, ChAd48, ChAd49, ChAd49, ChAd50, ChAd67, or SA7P), or rhesus adenoviruses. The viral vector can be used to infect cells of a subject, which, in turn, promotes the translation of the heterologous gene(s) of the viral vector into the immunogens of the invention. For example, a viral vector of the invention can be genetically modified to contain one or more nucleic acid sequences set forth in SEQ ID NOs: 1, 3, 5, 7, 9, or 11 or variants thereof having at least 85-99% sequence identity thereto, for example at least greater than 90% sequence identity thereto, and complements thereof.

[0061] Adenoviral vectors disclosed in International Patent Application Publications W02006/040330 and W02007/ 104792, each incorporated by reference herein, are particularly useful as vectors of the invention. These adenoviral vectors can encode and/or deliver one or more of the immunogens of the invention (e.g., ZIKV polypeptides) to treat a subject having a pathological condition associated with a viral infection (e.g., a ZIKV infection). In some embodiments, one or more recombinant adenovirus vectors can be administered to the subject in order to express more than one type of immunogen (e.g., ZIKV polypeptide) of the invention.

Adenoviruses

[0062] An adenovirus according to the invention belongs to the family of the

Adenoviridae and preferably is one that belongs to the genus Mastadenovirus. It can be a human adenovirus, but also an adenovirus that infects other species, including but not limited to a bovine adenovirus (e.g. bovine adenovirus 3, BAdV3), a canine adenovirus (e.g. CAdV2), a porcine adenovirus (e.g. PAdV3 or 5), or a simian adenovirus (which includes a monkey adenovirus and an ape adenovirus, such as a chimpanzee adenovirus or a gorilla adenovirus). Preferably, the adenovirus is a human adenovirus (HAdV, or AdHu; in the invention a human adenovirus is meant if referred to Ad without indication of species, e.g. the brief notation“Ad5” means the same as HAdV5, which is human adenovirus serotype 5), or a simian adenovirus such as chimpanzee or gorilla adenovirus (ChAd, AdCh, or SAdV).

[0063] Most advanced studies have been performed using human adenoviruses, and human adenoviruses are preferred according to certain aspects of the invention. In certain preferred embodiments, the recombinant adenovirus according to the invention is based upon a human adenovirus. In preferred embodiments, the recombinant adenovirus is based upon a human adenovirus serotype 5, 11, 26, 34, 35, 48, 49 or 50. According to a particularly preferred embodiment of the invention, an adenovirus is a human adenovirus of one of the serotypes 26 or 35.

[0064] An advantage of these serotypes is a low seroprevalence and/or low pre-existing neutralizing antibody titers in the human population. Preparation of rAd26 vectors is described, for example, in WO 2007/104792 and in Abbink et al., (2007) Virol

8l(9):4654-63, both of which are incorporated by reference herein in their entirety.

Exemplary genome sequences of Ad26 are found in GenBank Accession EF 153474 and in SEQ ID NO: l of WO 2007/104792. Preparation of rAd35 vectors is described, for example, in US Patent No. 7,270,811, in WO 00/70071, and in Vogels et al., (2003) J Virol 77(15): 8263-71, all of which are incorporated by reference herein in their entirety. Exemplary genome sequences of Ad35 are found in GenBank Accession AC 000019 and in Fig. 6 of WO 00/70071.

[0065] Simian adenoviruses generally also have a low seroprevalence and/or low pre- existing neutralizing antibody titers in the human population, and a significant amount of work has been reported using chimpanzee adenovirus vectors (e.g. US6083716; WO 2005/071093; WO 2010/086189; WO 2010085984; Farina et al, 2001, J Virol 75: 11603- 13; Cohen et al, 2002, J Gen Virol 83: 151-55; Kobinger et al, 2006, Virology 346: 394- 401; Tatsis et al., 2007, Molecular Therapy 15: 608-17; see also review by Bangari and Mittal, 2006, Vaccine 24: 849- 62; and review by Lasaro and Ertl, 2009, Mol Ther 17: 1333-39). Hence, in other preferred embodiments, the recombinant adenovirus according to the invention is based upon a simian adenovirus, e.g. a chimpanzee adenovirus. In certain embodiments, the recombinant adenovirus is based upon simian adenovirus type 1,

7, 8, 21, 22, 23, 24, 25, 26, 27.1, 28.1, 29, 30, 31.1, 32, 33, 34, 35.1, 36, 37.2, 39, 40.1,

41.1, 42.1, 43, 44, 45, 46, 48, 49, 50 or SA7P.

[0066] Adenoviral Vector rAd26

[0067] In a preferred embodiment according to the invention the adenoviral vectors comprise capsid proteins from two rare serotypes: Ad26 or Ad35. In the typical embodiment, the vector is an rAd26 virus.

[0068] Thus, the vectors that can be used in the invention comprise an Ad26 capsid protein (e.g., a fiber, penton or hexon protein). One of skill will recognize that it is not necessary that an entire Ad26 capsid protein be used in the vectors of the invention. Thus, chimeric capsid proteins that include at least a part of an Ad26 capsid protein can be used in the vectors of the invention. The vectors of the invention can also comprise capsid proteins in which the fiber, penton, and hexon proteins are each derived from a different serotype, so long as at least one capsid protein is derived from Ad26. In preferred embodiments, the fiber, penton and hexon proteins are each derived from Ad26.

[0069] One of skill will recognize that elements derived from multiple serotypes can be combined in a single recombinant adenovirus vector. Thus, a chimeric adenovirus that combines desirable properties from different serotypes can be produced. Thus, in some embodiments, a chimeric adenovirus of the invention can combine the absence of pre- existing immunity of the Ad26 serotypes with characteristics such as temperature stability, assembly, anchoring, production yield, redirected or improved infection, stability of the DNA in the target cell, and the like.

[0070] In certain embodiments the recombinant adenovirus vector useful in the invention is derived mainly or entirely from Ad26 (i.e., the vector is rAd26). In some embodiments, the adenovirus is replication deficient, e.g. because it contains a deletion in the El region of the genome. For the adenoviruses of the invention, being derived from Ad26, it is typical to exchange the E4-orf6 coding sequence of the adenovirus with the E4- orf6 of an adenovirus of human subgroup C such as Ad5. This allows propagation of such adenoviruses in well-known complementing cell lines that express the El genes of Ad5, such as for example 293 cells, PER.C6 cells, and the like (see, e.g. Havenga et al, 2006, J Gen Virol 87: 2135-43; WO 03/104467). In certain embodiments, the adenovirus is a human adenovirus of serotype 35, with a deletion in the El region into which the nucleic acid encoding the antigen has been cloned, and with an E4 orf6 region of Ad5. In certain embodiments, the adenovirus is a human adenovirus of serotype 26, with a deletion in the El region into which the nucleic acid encoding the antigen has been cloned, and with an E4 orf6 region of Ad5. For the Ad35 adenovirus, it is typical to retain the 3’ end of the E1B 55K open reading frame in the adenovirus, for instance the 166 bp directly upstream of the pIX open reading frame or a fragment comprising this such as a 243 bp fragment directly upstream of the pIX start codon, marked at the 5’ end by a Bsu36I restriction site, since this increases the stability of the adenovirus because the promoter of the pIX gene is partly residing in this area (see, e.g. Havenga et al, 2006, supra; WO 2004/001032). The preparation of recombinant adenoviral vectors is well known in the art.

[0071] Preparation of rAd26 vectors is described, for example, in WO 2007/104792 and in Abbink et al., (2007) Virol 81(9): 4654-63. Exemplary genome sequences of Ad26 are found in GenBank Accession EF 153474 and in SEQ ID NO:l of WO 2007/104792. Preparation of rAd35 vectors is described, for example, in ETS Patent No. 7,270,811 and in Vogels et al., (2003) J Virol 77(15): 8263-71. An exemplary genome sequence of Ad35 is found in GenBank Accession AC 000019.

[0072] In an embodiment of the invention, the vectors useful for the invention include those described in WO2012/082918, the disclosure of which is incorporated herein by reference in its entirety.

[0073] Typically, a vector useful in the invention is produced using a nucleic acid comprising the entire recombinant adenoviral genome (e.g., a plasmid, cosmid, or baculovirus vector). Thus, the invention also provides isolated nucleic acid molecules that encode the adenoviral vectors of the invention. The nucleic acid molecules of the invention can be in the form of RNA or in the form of DNA obtained by cloning or produced synthetically. The DNA can be double- stranded or single-stranded.

[0074] The adenovirus vectors useful in the invention are typically replication defective. In these embodiments, the virus is rendered replication-defective by deletion or inactivation of regions critical to replication of the virus, such as the El region. The regions can be substantially deleted or inactivated by, for example, inserting the gene of interest (usually linked to a promoter). In some embodiments, the vectors of the invention can contain deletions in other regions, such as the E2, E3 or E4 regions or insertions of heterologous genes linked to a promoter. For E2- and/or E4-mutated adenoviruses, generally E2- and/or E4-complementing cell lines are used to generate recombinant adenoviruses. Mutations in the E3 region of the adenovirus need not be complemented by the cell line, since E3 is not required for replication.

[0075] A packaging cell line is typically used to produce sufficient amount of adenovirus vectors of the invention. A packaging cell is a cell that comprises those genes that have been deleted or inactivated in a replication-defective vector, thus allowing the virus to replicate in the cell. Suitable cell lines include, for example, PER.C6, 911, 293, and El A549.

Pharmaceutical Compositions

[0076] In another general aspect, the invention relates to pharmaceutical compositions comprising adenoviral vectors (or adenoviral particles) comprising a nucleic acid molecule encoding a Zika virus antigen of the invention and a pharmaceutically acceptable carrier. Adenoviral vectors (or particles) of the invention and compositions comprising them are also useful in the manufacture of a medicament for therapeutic applications mentioned herein.

[0077] By“pharmaceutical composition” is meant any composition that contains a therapeutically or biologically active agent, such as an immunogenic composition or vaccine of the invention (e.g., an adenoviral particle comprising a ZIKV nucleic acid molecule and/or polypeptide of the invention), preferably including a nucleotide sequence encoding an antigenic gene product of interest, or fragment thereof, that is suitable for administration to a subject and that treats or prevents a disease (e.g., ZIKV infection) or reduces or ameliorates one or more symptoms of the disease (e.g., ZIKV viral titer, viral spread, infection, and/or cell fusion)). For the purposes of this invention, pharmaceutical compositions include vaccines, and pharmaceutical compositions suitable for delivering a therapeutic or biologically active agent can include, for example, tablets, gelcaps, capsules, pills, powders, granulates, suspensions, emulsions, solutions, gels, hydrogels, oral gels, pastes, eye drops, ointments, creams, plasters, drenches, delivery devices, suppositories, enemas, injectables, implants, sprays, or aerosols. Any of these formulations can be prepared by well-known and accepted methods of art. See, for example, Remington: The Science and Practice of Pharmacy (2 I st ed.), ed. A.R. Gennaro, Lippincott Williams & Wilkins, 2005, and Encyclopedia of Pharmaceutical Technology , ed. J. Swarbrick, Informa Healthcare, 2006, each of which is hereby incorporated by reference.

[0078] As used herein, the term“carrier” refers to any excipient, diluent, filler, salt, buffer, stabilizer, solubilizer, oil, lipid, lipid containing vesicle, microsphere, liposomal encapsulation, or other material well known in the art for use in pharmaceutical formulations. It will be understood that the characteristics of the carrier, excipient or diluent will depend on the route of administration for a particular application. As used herein, the term“pharmaceutically acceptable carrier” refers to a non-toxic material that does not interfere with the effectiveness of a composition according to the invention or the biological activity of a composition according to the invention. According to particular embodiments, in view of the present disclosure, any pharmaceutically acceptable carrier suitable for use in a pharmaceutical composition can be used in the invention.

[0079] Pharmaceutically acceptable acidic/anionic salts for use in the invention include, and are not limited to acetate, benzenesulfonate, benzoate, bicarbonate, bitartrate, bromide, calcium edetate, camsylate, carbonate, chloride, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, fumarate, glyceptate, gluconate, glutamate, glycollylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, malate, maleate, mandelate, mesylate, methylbromide, methylnitrate, methyl sulfate, mucate, napsylate, nitrate, pamoate, pantothenate, phosphate/diphosphate, polygalacturonate, salicylate, stearate, subacetate, succinate, sulfate, tannate, tartrate, teoclate, tosylate and triethiodide.

Organic or inorganic acids also include, and are not limited to, hydriodic, perchloric, sulfuric, phosphoric, propionic, glycolic, methanesulfonic, hydroxyethanesulfonic, oxalic, 2-naphthalenesulfonic, p-toluenesulfonic, cyclohexanesulfamic, saccharinic or trifluoroacetic acid.

[0080] Pharmaceutically acceptable basic/cationic salts include, and are not limited to aluminum, 2-amino-2-hydroxymethyl-propane-l,3-diol (also known as

tris(hydroxymethyl)aminomethane, tromethane or“TRIS”), ammonia, benzathine, t-butylamine, calcium, chloroprocaine, choline, cyclohexylamine, diethanolamine, ethylenediamine, lithium, L-lysine, magnesium, meglumine, N-methyl-D-glucamine, piperidine, potassium, procaine, quinine, sodium, triethanolamine, or zinc.

[0081] In some embodiments of the invention, pharmaceutical compositions are provided comprising the adenoviral vectors of the invention in an amount from about 1 x 10 10 , about 2 x 10 10 , about 3 x 10 10 , about 4 x 10 10 , about 5 x 10 10 , about 6 x 10 10 , about 7 x 10 10 , about 8 x 10 10 , about 9 x 10 10 , about 1 x 10 11 , about 2 x 10 11 , about 3 x 10 11 , about 4 x 10 11 , or about 5 x 10 11 viral vectors (or particles) per dose. In certain embodiments of the invention, the pharmaceutical composition comprises about 1 x 10 10 adenoviral vectors (or particles) to about 5 x 10 11 adenoviral vectors (or particles) per dose. In certain embodiments of the invention, the pharmaceutical composition comprises about 5 x 10 10 adenoviral vectors (or particles) to about 1 x 10 11 adenoviral vectors (or particles) per dose. In certain embodiments of the invention, the pharmaceutical composition comprises about 5 x 10 10 adenoviral vectors (or particles) per dose. In certain

embodiments of the invention, the pharmaceutical composition comprises about 1 x 10 11 adenoviral vectors (or particles) per dose.

[0082] The pharmaceutical composition can have a pH from about 3.0 to about 10, for example from about 3 to about 7, or from about 5 to about 9. The formulation can further comprise at least one ingredient selected from the group consisting of a buffer system, preservative(s), tonicity agent(s), chelating agent(s), stabilizer(s) and surfactant(s).

[0083] In certain embodiments, the subject is administered a single dose of the pharmaceutical composition. In certain embodiments, the subject is administered a double dose of the pharmaceutical composition. When administering a double dose, the first and second dose of the pharmaceutical composition can be administered to the subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart.

[0084] The formulation of pharmaceutically active ingredients with pharmaceutically acceptable carriers is known in the art, e.g., Remington: The Science and Practice of Pharmacy (e.g. 2lst edition (2005), and any later editions). Non-limiting examples of additional ingredients include: buffers, diluents, solvents, tonicity regulating agents, preservatives, stabilizers, and chelating agents. One or more pharmaceutically acceptable carriers can be used in formulating the pharmaceutical compositions of the invention.

[0085] In one embodiment of the invention, the pharmaceutical composition is a liquid formulation. A preferred example of a liquid formulation is an aqueous formulation, i.e., a formulation comprising water. The liquid formulation can comprise a solution, a suspension, an emulsion, a microemulsion, a gel, and the like. An aqueous formulation typically comprises at least 50% w/w water, or at least 60%, 70%, 75%, 80%, 85%, 90%, or at least 95% w/w of water.

[0086] In one embodiment, the pharmaceutical composition can be formulated as an injectable which can be injected, for example, via a syringe or an infusion pump. The injection can be delivered subcutaneously, intramuscularly, intraperitoneally, or intravenously, for example.

[0087] In another embodiment, the pharmaceutical composition is a solid formulation, e.g., a freeze-dried or spray-dried composition, which can be used as is, or whereto the physician or the patient adds solvents, and/or diluents prior to use. Solid dosage forms can include tablets, such as compressed tablets, and/or coated tablets, and capsules (e.g., hard or soft gelatin capsules). The pharmaceutical composition can also be in the form of sachets, dragees, powders, granules, lozenges, or powders for reconstitution, for example.

[0088] The dosage forms can be immediate release, in which case they can comprise a water-soluble or dispersible carrier, or they may be delayed release, sustained release, or modified release, in which case they may comprise water-insoluble polymers that regulate the rate of dissolution of the dosage form in the gastrointestinal tract.

[0089] In other embodiments, the pharmaceutical composition can be delivered intranasally, intrabuccally, or sublingually.

[0090] The pH in an aqueous formulation can be between pH 3 and pH 10. In one embodiment of the invention, the pH of the formulation is from about 7.0 to about 9.5. In another embodiment of the invention, the pH of the formulation is from about 3.0 to about 7.0.

[0091] In another embodiment of the invention, the pharmaceutical composition comprises a buffer. Non-limiting examples of buffers include: arginine, aspartic acid, bicine, citrate, disodium hydrogen phosphate, fumaric acid, glycine, glycylglycine, histidine, lysine, maleic acid, malic acid, sodium acetate, sodium carbonate, sodium dihydrogen phosphate, sodium phosphate, succinate, tartaric acid, tricine, and

tris(hydroxymethyl)-aminomethane, and mixtures thereof. The buffer may be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific buffers constitute alternative embodiments of the invention.

[0092] In another embodiment of the invention, the pharmaceutical composition comprises a preservative. Non-limiting examples of preservatives include: benzethonium chloride, benzoic acid, benzyl alcohol, bronopol, butyl 4-hydroxybenzoate,

chlorobutanol, chlorocresol, chlorohexidine, chlorphenesin, o-cresol, m-cresol, p-cresol, ethyl 4-hydroxybenzoate, imidurea, methyl 4-hydroxybenzoate, phenol, 2- phenoxyethanol, 2-phenylethanol, propyl 4-hydroxybenzoate, sodium dehydroacetate, thiomerosal, and mixtures thereof. The preservative may be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific preservatives constitute alternative embodiments of the invention.

[0093] In another embodiment of the invention, the pharmaceutical composition comprises an isotonic agent. Non-limiting examples of isotonic agents include a salt (such as sodium chloride), an amino acid (such as glycine, histidine, arginine, lysine, isoleucine, aspartic acid, tryptophan, and threonine), an alditol (such as glycerol, 1,2- propanediol propyleneglycol), 1,3 -propanediol, and l,3-butanediol), poly ethyleneglycol (e.g. PEG400), and mixtures thereof. Another example of an isotonic agent includes a sugar. Non-limiting examples of sugars may be mono-, di-, or polysaccharides, or water- soluble glucans, including for example fructose, glucose, mannose, sorbose, xylose, maltose, lactose, sucrose, trehalose, dextran, pullulan, dextrin, cyclodextrin, alpha and beta- HPCD, soluble starch, hydroxyethyl starch, and sodium carboxymethylcellulose. Another example of an isotonic agent is a sugar alcohol, wherein the term“sugar alcohol” is defined as a C(4-8) hydrocarbon having at least one— OH group. Non limiting examples of sugar alcohols include mannitol, sorbitol, inositol, galactitol, dulcitol, xylitol, and arabitol. Pharmaceutical compositions comprising each isotonic agent listed in this paragraph constitute alternative embodiments of the invention. The isotonic agent can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific isotonic agents constitute alternative embodiments of the invention.

[0094] In another embodiment of the invention, the pharmaceutical composition comprises a chelating agent. Non-limiting examples of chelating agents include citric acid, aspartic acid, salts of ethylenediaminetetraacetic acid (EDTA), and mixtures thereof. The chelating agent can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific chelating agents constitute alternative embodiments of the invention.

[0095] In another embodiment of the invention, the pharmaceutical composition comprises a stabilizer. Non-limiting examples of stabilizers include one or more aggregation inhibitors, one or more oxidation inhibitors, one or more surfactants, and/or one or more protease inhibitors.

[0096] In another embodiment of the invention, the pharmaceutical composition comprises a stabilizer, wherein said stabilizer is carboxy-/hydroxycellulose and derivates thereof (such as HPC, HPC-SL, HPC-L and HPMC), cyclodextrins, 2-methylthioethanol, polyethylene glycol (such as PEG 3350), polyvinyl alcohol (PVA), polyvinyl

pyrrolidone, salts (such as sodium chloride), sulphur-containing substances such as monothioglycerol), or thioglycolic acid. The stabilizer can be present individually or in the aggregate, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific stabilizers constitute alternative embodiments of the invention.

[0097] In further embodiments of the invention, the pharmaceutical composition comprises one or more surfactants, preferably a surfactant, at least one surfactant, or two different surfactants. The term“surfactant” refers to any molecules or ions that are comprised of a water-soluble (hydrophilic) part, and a fat-soluble (lipophilic) part. The surfactant can, for example, be selected from the group consisting of anionic surfactants, cationic surfactants, nonionic surfactants, and/or zwitterionic surfactants. The surfactant can be present individually or in the aggregate, in a concentration from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific surfactants constitute alternative embodiments of the invention.

[0098] In a further embodiment of the invention, the pharmaceutical composition comprises one or more protease inhibitors, such as, e.g., EDTA (ethylenediamine tetraacetic acid), and/or benzamidine hydrochloric acid (HC1). The protease inhibitor can be present individually or in the aggregate, in a concentration from about 0.1 mg/ml to about 20 mg/ml. Pharmaceutical compositions comprising each one of these specific protease inhibitors constitute alternative embodiments of the invention.

[0099] The pharmaceutical composition of the invention can comprise an amount of an amino acid base sufficient to decrease aggregate formation of the polypeptide during storage of the composition. The term“amino acid base” refers to one or more amino acids (such as methionine, histidine, imidazole, arginine, lysine, isoleucine, aspartic acid, tryptophan, threonine), or analogues thereof. Any amino acid can be present either in its free base form or in its salt form. Any stereoisomer (i.e., L, D, or a mixture thereof) of the amino acid base may be present. The amino acid base can be present individually or in the combination with other amino acid bases, in a concentration from about 0.01 mg/ml to about 50 mg/ml, for example from about 0.1 mg/ml to about 20 mg/ml.

Pharmaceutical compositions comprising each one of these specific amino acid bases constitute alternative embodiments of the invention.

[00100] It is also apparent to one skilled in the art that the therapeutically effective dose for adenoviral particles comprising a nucleic acid molecule encoding a Zika virus antigen of the present invention or a pharmaceutical composition thereof will vary according to the desired effect. Therefore, optimal dosages to be administered can be readily determined by one skilled in the art and will vary with the particular adenoviral particle used, the mode of administration, the strength of the preparation, and the advancement of the disease condition (e.g., Zika virus infection). In addition, factors associated with the particular subject being treated, including subject age, weight, diet and time of administration, will result in the need to adjust the dose to an appropriate therapeutic level. [00101] The pharmaceutically-acceptable salts of the adenoviral particles of the invention include the conventional non-toxic salts or the quaternary ammonium salts which are formed from inorganic or organic acids or bases. Examples of such acid addition salts include acetate, adipate, benzoate, benzenesulfonate, citrate, camphorate, dodecyl sulfate, hydrochloride, hydrobromide, lactate, maleate, methanesulfonate, nitrate, oxalate, pivalate, propionate, succinate, sulfate and tartrate. Base salts include ammonium salts, alkali metal salts such as sodium and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, salts with organic bases such as dicyclohexylamino salts and salts with amino acids such as arginine. Also, the basic nitrogen-containing groups may be quaternized with, for example, alkyl halides.

[00102] The pharmaceutical compositions of the invention can be administered by any means that accomplish their intended purpose. As used herein, by“administering” is meant a method of giving a dosage of a pharmaceutical composition (e.g., an

immunogenic composition (e.g., a vaccine (e.g., a Zika virus (ZIKV) vaccine))) to a subject. The compositions utilized in the methods described herein can be administered, for example, intramuscularly, intravenously, intradermally, percutaneously,

intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally,

intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, by catheter, by lavage, by gavage, in cremes, or in lipid compositions. The preferred method of administration can vary depending on various factors (e.g., the components of the composition being administered and the severity of the condition being treated).

Methods of use

[00103] The present invention provides methods for generating an immune response against a Zika virus in a human subject in need thereof. The methods comprise administering to the subject a pharmaceutical composition comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier. The methods are for preventing, treating, delaying the onset of, or ameliorating a Zika virus infection or any one or more symptoms of said Zika virus infection, the method comprising administering to the subject in need thereof an effective amount of a pharmaceutical composition of the invention.

[00104] According to particular embodiments, an immunogenic or effective or protective amount refers to the amount of an immunogen which is sufficient to achieve one, two, three, four, or more of the following effects: (i) reduce or ameliorate the severity of the Zika virus infection to be treated or a symptom associated therewith; (ii) reduce the duration of the Zika virus infection to be treated, or a symptom associated therewith; (iii) prevent the progression of the Zika virus infection to be treated, or a symptom associated therewith; (iv) cause regression of the Zika virus infection to be treated, or a symptom associated therewith; (v) prevent the development or onset of the Zika virus infection to be treated, or a symptom associated therewith; (vi) prevent the recurrence of the Zika virus infection to be treated, or a symptom associated therewith; (vii) reduce hospitalization of a subject having the Zika virus infection or condition to be treated, or a symptom associated therewith; (viii) reduce hospitalization length of a subject having the Zika virus infection to be treated, or a symptom associated therewith; (ix) increase the survival of a subject with the Zika virus infection to be treated, or a symptom associated therewith; (xi) inhibit or reduce the Zika virus infection to be treated, or a symptom associated therewith in a subject; and/or (xii) enhance or improve the prophylactic or therapeutic effect(s) of another therapy; (xiii) prevent transmission of zika virus through sexual and maternal to fetal routes; (xiv) prevent and/or reduces the severity of fetal brain abnormalities associated with Zika virus.

[00105] Examples of symptoms of diseases caused by a viral infection, such as ZIKV, that can be prevented using the compositions of the invention include, for example, fever, joint pain, rash, conjunctivitis, muscle pain, headache, retro-orbital pain, edema, lymphadenopathy, malaise, asthenia, sore throat, cough, nausea, vomiting, diarrhea, and hematospermia. These symptoms, and their resolution during treatment, can be measured by, for example, a physician during a physical examination or by other tests and methods known in the art.

[00106] The immunogenic or effective amount or dosage can vary according to various factors, such as the Zika virus infection to be treated, the means of administration, the target site, the physiological state of the subject (including, e.g., age, body weight, health), whether the subject is a human or an animal, other medications administered, and whether the treatment is prophylactic or therapeutic. Treatment dosages are optimally titrated to optimize safety and efficacy.

[00107] As used herein, the terms“treat,”“treating,” and“treatment” are all intended to refer to an amelioration or reversal of at least one measurable physical parameter related to the Zika virus infection, which is not necessarily discernible in the subject, but can be discernible in the subject. The terms“treat,”“treating,” and“treatment,” can also refer to causing regression, preventing the progression, or at least slowing down the progression of the Zika virus infection. In a particular embodiment,“treat,”“treating,” and “treatment” refer to an alleviation, prevention of the development or onset, or reduction in the duration of one or more symptoms associated with the Zika virus infection. In a particular embodiment,“treat,”“treating,” and“treatment” refer to prevention of the recurrence of the Zika virus infection. In a particular embodiment,“treat,”“treating,” and“treatment” refer to an increase in the survival of a subject having the Zika virus infection. In a particular embodiment,“treat,”“treating,” and“treatment” refer to elimination of the Zika virus infection in the subject.

[00108] In certain embodiments, administration of an immunogenic or effective amount of a pharmaceutical composition of the invention reduces ZIKV serum viral loads determined from a subject having a ZIKV infection by at least about 5%, 10%, 20%,

30%, 40%, 50%, 60%, 70%, 80%, 90%, or more compared to viral loads determined from the subject prior to administration of an effective amount of a composition of the invention. In some instances, administration of an effective amount of a composition of the invention reduces serum viral loads to an undetectable level compared to viral loads determined from the subject prior to administration of an effective amount of a composition of the invention. In some instances, administration of an effective amount of a composition of the invention results in a reduced and/or undetectable serum viral load that may be maintained for at least about 1, 2, 3, 4, 5, 6, 7 days; 1, 2, 3, 4, weeks; 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months; or 1 year or more.

[00109] In addition, single or multiple administrations of the compositions of the present invention can be given (pre- or post-exposure and/or pre- or post-diagnosis) to a subject (e.g., one administration or administration two or more times). For example, subjects who are particularly susceptible to, for example, viral infection (e.g., a ZIKV infection) can require multiple administrations of the compositions of the present invention to establish and/or maintain protection against the virus. Levels of induced immunity provided by the pharmaceutical compositions described herein can be monitored by, for example, measuring amounts of neutralizing secretory and serum antibodies. The dosages can then be adjusted or repeated as necessary to trigger the desired level of immune response. For example, the immune response triggered by a single administration (prime) of a composition of the invention may not sufficiently potent and/or persistent to provide effective protection. Accordingly, in some embodiments, repeated administration (boost), such that a prime boost regimen is established, can significantly enhance humoral and cellular responses to the antigen of the composition.

[00110] Alternatively, the efficacy of treatment can be determined by monitoring the level of the antigenic or therapeutic gene product, or fragment thereof, expressed in a subject (e.g., a human) following administration of the pharmaceutical compositions of the invention. For example, the blood or lymph of a subject can be tested for antigenic or therapeutic gene product, or fragment thereof, using, for example, standard assays known in the art.

[00111] In some instances, efficacy of treatment can be determined by monitoring a change in the serum viral load from a sample from the subject obtained prior to and after administration of an effective amount of a pharmaceutical composition of the invention.

A reduction in serum viral load of at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or more compared to viral load determined from the subject prior to administration of an effective amount of a composition of the invention can indicate that the subject is receiving benefit from the treatment. If a viral load does not decrease by at least about 10%, 20%, 30%, or more after administration of a composition of the invention, the dosage of the composition to be administered can be increased. For example, by increasing the number of viral particles (VP) of an adenovirus vector-based vaccine. [00112] Immunogenicity of the pharmaceutical compositions of the invention can be improved if it is co-administered with an immunostimulatory agent and/or adjuvant. Suitable adjuvants well-known to those skilled in the art include, for example, aluminum phosphate, aluminum hydroxide, QS21, Quil A (and derivatives and components thereof), calcium phosphate, calcium hydroxide, zinc hydroxide, glycolipid analogs, octodecyl esters of an amino acid, muramyl dipeptides, polyphosphazene, lipoproteins, ISCOM matrix, DC-Chol, DDA, cytokines, and other adjuvants and derivatives thereof.

[00113] The term“immunostimulatory agenf‘ refers to substances (e.g., drugs and nutrients) that stimulate the immune system by inducing activation or increasing activity of any of its components. An immunostimulatory agent can, for example, include a cytokine (e.g., the granulocyte macrophage colony-stimulating factor) and interferon (e.g., IFN-a and/or IFN-g).

[00114] The term“adjuvant” is defined as a pharmacological or immunological agent that modifies the effect of other agents (e.g., a ZIKV antigen) while having few if any direct effects when administered alone. An adjuvant can be one or more substances that cause stimulation of the immune system. In this context, an adjuvant is used to enhance an immune response to the adenoviral particles of the invention.

EMBODIMENTS

[00115] The invention provides also the following non-limiting embodiments.

[00116] Embodiment l is a method for generating an immune response against a Zika virus in a human subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier, wherein about 5 x 10 10 adenoviral vectors (or particles) to about 1 x 10 11 adenoviral vectors (or particles) are administered per dose to the subject in need thereof.

[00117] Embodiment 2 is the method of embodiment 1, for generating a protective immune response against a Zika virus in a human subject in need thereof.

[00118] Embodiment 3 is the method of embodiment 1 or 2, wherein the

pharmaceutical composition is administered via an intramuscular injection to the human subject in need thereof. [00119] Embodiment 4 is the method of any one of embodiments 1-3, wherein the pharmaceutical composition is administered to the human subject as a single dose.

[00120] Embodiment 5 is the method of any one of embodiments 1-3, wherein the pharmaceutical composition is administered to the human subject as a double dose.

[00121] Embodiment 6 is the method of embodiment 5, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart.

[00122] Embodiment 7 is the method of embodiment 5, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about eight weeks apart.

[00123] Embodiment 8 is the method of any one of embodiments 1-7, wherein about 5 x 10 10 , about 6 x 10 10 , about 7 x 10 10 , about 8 x 10 10 , about 9 x 10 10 , or about 1 x 10 11 of the adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00124] Embodiment 9 is the method of embodiment 8, wherein about 5 x 10 10 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00125] Embodiment 10 is the method of embodiment 8, wherein about 1 x 10 11 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00126] Embodiment 11 is the method of any one of embodiments 1-10, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2, 4, 6, 8, 10, or 12.

[00127] Embodiment 12 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2.

[00128] Embodiment 13 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:4.

[00129] Embodiment 14 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:6.

[00130] Embodiment 15 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:8.

[00131] Embodiment 16 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 10. [00132] Embodiment 17 is the method of embodiment 11, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 12.

[00133] Embodiment 18 is the method of any one of embodiments 1-17, wherein the adenoviral vector serotype is selected from the group consisting of Ad2, Ad5, Adl 1,

Ad 12, Ad24, Ad26, Ad34, Ad35, Ad40, Ad48, Ad49, Ad50, Ad52, and Pan9.

[00134] Embodiment 19 is the method of embodiment 18, wherein the adenoviral vector serotype is the Ad26 serotype.

[00135] Embodiment 20 is a method for preventing a Zika virus infection or the progression of a Zika virus infection in a human subject in need thereof, the method comprising administering to the subject a pharmaceutical composition comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier, wherein the pharmaceutical composition comprises a total dosage of about 5 x 10 10 adenoviral vectors (or particles) to about 1 x 10 11 adenoviral vectors (or particles) per administration.

[00136] Embodiment 21 is the method of embodiment 20, wherein the pharmaceutical composition is administered intramuscularly, intravenously, intradermally,

percutaneously, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally,

intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, by catheter, by lavage, or by gavage.

[00137] Embodiment 22 is the method of embodiment 21, wherein the pharmaceutical composition is administered in an intramuscular injection.

[00138] Embodiment 23 is the method of any one of embodiments 20-22, wherein the pharmaceutical composition is administered as a single dose.

[00139] Embodiment 24 is the method of any one of embodiments 20-22, wherein the pharmaceutical composition is administered as a double dose.

[00140] Embodiment 25 is the method of embodiment 24, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart.

[00141] Embodiment 26 is the method of embodiment 25, wherein the first and second dose of the pharmaceutical composition are administered to the human subject about eight weeks apart.

[00142] Embodiment 27 is the method of any one of the embodiments 20-26, wherein about 5 x 10 10 , about 6 x 10 10 , about 7 x 10 10 , about 8 x 10 10 , about 9 x 10 10 , or about 1 x 10 11 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00143] Embodiment 28 is the method of embodiment 27, wherein about 5 x 10 10 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00144] Embodiment 29 is the method of embodiment 27, wherein about 1 x 10 11 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00145] Embodiment 30 is the method of any one of embodiments 20-29, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2, 4, 6, 8, 10, or 12.

[00146] Embodiment 31 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2.

[00147] Embodiment 32 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:4.

[00148] Embodiment 33 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:6.

[00149] Embodiment 34 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:8.

[00150] Embodiment 35 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 10.

[00151] Embodiment 36 is the method of embodiment 30, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 12.

[00152] Embodiment 37 is the method of any one of embodiments 20-36, wherein the adenoviral vector serotype is selected from the group consisting of Ad2, Ad5, Adl 1,

Ad 12, Ad24, Ad26, Ad34, Ad35, Ad40, Ad48, Ad49, Ad50, Ad52, and Pan9. [00153] Embodiment 38 is the method of embodiment 37, wherein the adenoviral vector serotype is the Ad26 serotype.

[00154] Embodiment 39 is the use of a pharmaceutical composition for the preparation of a vaccine for generating an immune response against a Zika virus in a human subject in need thereof, comprising adenoviral vectors comprising a nucleic acid sequence encoding a Zika virus antigen and a pharmaceutically acceptable carrier, wherein about 5 x 10 10 adenoviral vectors (or particles) to about 1 x 10 11 adenoviral vectors (or particles) are administered per dose to the subject in need thereof.

[00155] Embodiment 40 is the use of embodiment 39, wherein the vaccine is administered intramuscularly, intravenously, intradermally, percutaneously,

intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally,

intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctivally, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularly, orally, topically, locally, by inhalation, by injection, by infusion, by continuous infusion, by localized perfusion, by catheter, by lavage, or by gavage.

[00156] Embodiment 41 is the use of embodiment 40, wherein the vaccine is administered in an intramuscular injection.

[00157] Embodiment 42 is the use of any one of embodiments 39-41, wherein the vaccine is administered as a single dose.

[00158] Embodiment 43 is the use of any one of embodiments 39-41, wherein the vaccine is administered as a double dose.

[00159] Embodiment 44 is the use of embodiment 43, wherein the first and second dose of the vaccine are administered to the human subject about four weeks, about eight weeks, about twelve weeks, about three months, about six months, about nine months, about one year, or about two years apart.

[00160] Embodiment 45 is the use of embodiment 44, wherein the first and second dose of the vaccine are administered to the human subject about eight weeks apart.

[00161] Embodiment 46 is the use of any one of the embodiments 39-45, wherein about 5 x 10 10 , about 6 x 10 10 , about 7 x 10 10 , about 8 x 10 10 , about 9 x 10 10 , or about 1 x 10 11 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00162] Embodiment 47 is the use of embodiment 46, wherein about 5 x 10 10 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00163] Embodiment 48 is the use of embodiment 46, wherein about 1 x 10 11 adenoviral vectors (or particles) are administered to the human subject in need thereof.

[00164] Embodiment 49 is the use of any one of embodiments 39-48, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2, 4, 6, 8, 10, or 12.

[00165] Embodiment 50 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:2.

[00166] Embodiment 51 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:4.

[00167] Embodiment 52 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:6.

[00168] Embodiment 53 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO:8.

[00169] Embodiment 54 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 10.

[00170] Embodiment 55 is the use of embodiment 49, wherein the Zika virus antigen comprises the amino acid sequence of SEQ ID NO: 12.

[00171] Embodiment 56 is the use of any one of embodiments 39-55, wherein the adenoviral vector serotype is selected from the group consisting of Ad2, Ad5, Adl 1,

Ad 12, Ad24, Ad26, Ad34, Ad35, Ad40, Ad48, Ad49, Ad50, Ad52, and Pan9.

[00172] Embodiment 57 is the use of embodiment 56, wherein the adenoviral vector serotype is the Ad26 serotype.

EXAMPLES

[00173] Example 1: Clinical Study to Evaluate Safety, Tolerability, and

Immunogenicity of Intramuscularly Administered Adenoviral Particles Comprising a Zika Virus Antigen [00174] A clinical study was conducted for evaluating the safety, tolerability and immunogenicity of Ad26.ZIKV.00l vectors at a dose of 5xl0 10 viral particles (vp) or lxlO 11 vp, which were administered intramuscularly as single dose or in a two dose schedule in healthy adults.

[00175] The study was a randomized, placebo-controlled, double-blind first in human phase 1 study conducted in 100 healthy adult male and female subjects (> 18 to < 50 years of age). The study consists of a vaccination period in which subjects were administered the Ad26.ZIKV.00l vaccine at either a 5xl0 10 vp or lxlO 11 vp dose at baseline (Day 1) followed by a second administration on Day 57 of either a placebo or the Ad26.ZIKV.00l vaccine at a similar dose as the first administration.

[00176] Subjects were enrolled into 5 different groups of 20 healthy subjects each. Overall, subjects were randomized in parallel in 1 : 1 : 1 : 1 : 1 ratio to 1 of 5 vaccination groups to receive active vaccine or placebo (0.9% saline) through intramuscular (IM) injections (0.5 ml) as follows:

Group 1 : Ad26.ZIKV.00l (5xl0 10 vp) on Day 1, followed by a second administration of Ad26.ZIKV.00l (5xl0 10 vp) on Day 57, or

Group 2: Ad26.ZIKV.00l (5xl0 10 vp) on Day 1, followed by a Placebo on Day 57.

Group 3 : Ad26.ZIKV.00l (lxlO 11 vp) on Day 1, followed by a second administration of Ad26.ZIKV.00l (lxlO 11 vp) on Day 57, or

Group 4: Ad26.ZIKV.00l (lxlO 11 vp) on Day 1, followed by a Placebo on Day 57.

Group 5 : Placebo on Day 1, followed by a Placebo on Day 57.

The exemplary study vaccination schedules are summarized in Table 1.

[00177] Table 1 : Study Vaccination Schedules Group Immunization 1 Immunization 2 Immunization Group Size (Dose 1) (Dose 2) _ Schedule (Weeks) _ N

1 Ad26.ZIKV.00l Ad26.ZIKV.00l 0 - 8 20

(5xl0 10 vp) (5xl0 10 vp)

2 Ad26.ZIKV.00l Placebo 0 - 8 20

(5xl0 10 vp)

3 Ad26.ZIKV.00l Ad26.ZIKV.00l 0 - 8 20

(lxlO 11 vp) (lxlO 11 vp)

4 Ad26.ZIKV.00l Placebo 0 - 8 20

(lxlO 11 vp)

5 Placebo Placebo 0 - 8 20

Abbreviations: vp: viral particles; N: number of subjects to receive study vaccine

[00178] Vaccine Material

[00179] Ad26.ZIKV.00l is a monovalent vaccine composed of a recombinant, replication-incompetent adenovirus serotype 26 (Ad26) vector, constructed to encode the Zika virus (ZIKV) membrane (M) and envelope (Env) proteins (M-Env) (SEQ ID NO:2) derived from the ZIKV strain from Brazil (ZIKV-BR) BeH815744 strain.

[00180] Safety Endpoint

[00181] Safety was assessed by a collection of solicited local and systemic adverse events, unsolicited adverse events, serious adverse events, and immediately reportable events. Other safety assessments included clinical laboratory tests (hematology, biochemistry), vital signs measurements (heart rate, supine systolic and diastolic blood pressure, body temperature), and physical examinations at multiple time points.

[00182] Immunogenicitv endpoints

[00183] Blood samples for immunogenicity assessments (humoral and cellular immune responses) were collected at different time points. Blood samples (serum) for assessment of humoral immune responses were obtained from all subjects. Blood samples (PBMC) for assessment of cellular immune responses were obtained from a subset of subjects only.

[00184] Humoral and cellular immunogenicity assays can include, but are not limited to, the assays summarized in Tables 2 and 3.

[00185] Table 2: Summary of Humoral Immunogenicity Assays Assay _ Purpose

Secondary endpoints

ZIKV neutralization (VNA) Analysis of neutralizing antibodies to the vaccine strain (or other strain)

Exploratory endpoints

ZIKV antibody (ELISA) Analysis of antibodies binding to ZIKV, or

individual ZIKV proteins (e.g., Env-protein, M- protein)

ZIKV neutralization Analysis of neutralizing antibodies to vaccine strain (neutralization assay) (or other strain), as measured by an alternative

neutralization assay (different from the VNA used for the secondary endpoint)

Flavivirus neutralization (VNA) Analysis of neutralizing antibodies to flaviviruses other than ZIKV, such as YFV, JEV, WNV, DENV, and TBEV

ELISA: enzyme-linked immunosorbent assay

[00186] Table 3 : Summary of Cellular Immunogenicity Assays

[00187] Immunogenicity was assessed in a subset of 75 subjects at baseline (Day 1), at 14 days and 28 days after the first vaccination (Days 15 and 29), at the day of the boost vaccination (pre-dose, D57), and 14 days and 28 days after the boost vaccination (D71 and 85) using a virus neutralization assay (VNA) to analyze the neutralizing antibody response against ZIKV (Zika Microneutralization Assay (MN50)). This in vitro assay determines the capacity of vaccine-induced antibodies to prevent infection of a cell line by live ZIKV (Modjarrad et ak, The Lancet, Vol. 391, issue 10120, 10-16 February 2018, p. 563-571). Results from the remaining 25 subjects, from any other immunological assessment measuring response against ZIKV, or from further time points in the ongoing study were not yet available.

[00188] The actual values, geometric mean titers (GMT) and percentage of responders are shown in FIGS 1 A and B (Zika Microneutralization Assay (MN50). Table 4 shows the GMTs, the percentage of subjects with MN50 titers above 10 (lower limit of quantification of the MN50 VNA and assay positivity criterium) and 100 MN50 units, respectively, and the geometric fold increase for the Zika microneutralization assay.

Titers above 100 MN50 units have been associated with protection against ZIKV challenge in mouse and NHP models (Modjarrad et ah, The Lancet, Vol. 391, issue 10120, 10-16 February 2018, p. 563-571; Abbink et ak, Science, 09 Sep 2016, Vol. 353, Issue 6304, pp. 1129-1132; Abbink et ak, Sci Transl Med. 2017, Dec 13, 9(420)).

[00189] At baseline all subjects had a geometric mean titer below LLOQ. None of the placebo recipients developed Zika MN50 titers above the LLOQ of 10 at any time point post vaccination. Fifteen days post dose 1, a majority of vaccinated subjects developed ZIKV neutralizing antibody titers, irrespective of the dose given, and geometric mean titers increased to levels above 100 MN50 units in all groups by day 28. Specifically, geometric mean titers at Day 15 after the first vaccination were 58.8 and 29.8

respectively for the groups receiving Ad26.ZIKV.00l at 5xl0 10 viral particles (vp), resulting in a seroconversion rate (i.e. subjects with MN50 titers >10) of 80% (12/15) and 53% (8/15), and 53.5 and 52.9 for the groups that received Ad26.ZIKV.00l at lxlO 11 vp, resulting in a seroconversion rate of 78.6% (11/14) and 66.7% (10/15), respectively. Geometric mean titers at Day 29 after the first vaccination increased to 121.4 and 139.4 respectively for the groups receiving Ad26.ZIKV.00l at 5xl0 10 vp, resulting in seroconversion rates of 93.3% (14/15) and 86.7% (13/15), and 125 and 169 for the groups that received Ad26.ZIKV.00l at lxlO 11 vp, resulting in seroconversion rates of 92.2% (13/14) for both groups. The percentage of subjects whose MN50 titers exceeded 100 - a level that showed protection against ZIKV challenge in mouse and NHP models - increased from 36.7% (11/30) on day 15 to 56.7% (17/30) on day 28 in the combined groups receiving 5xl0 10 vp of Ad26.ZIKV.00l. In the combined groups that received Ad26.ZIKV.00l at lxlO 11 vp, 13/29 (46.7%) and 16/28 (58.6%) subjects reached MN50 titers of >100 at 14 days and 28 days after prime vaccination, respectively. [00190] Geometric mean titers at Day 57, prior to the second dose of either vaccine or placebo, were 37.5 and 64.9 respectively for the groups receiving 5xl0 10 vp of

Ad26.ZIKV.00l and slightly higher, 127 and 120.3, for the groups that received lxlO 11 vp of Ad26.ZIKV.00l as prime. In the group that received placebo after prime with 5xl0 10 vp of Ad26.ZIKV.00l, geometric mean titers remained at 64.2, 14 days after the second vaccination (day 71) and decreased slightly further to 44.8, 28 days after the second vaccination (day 85).

[00191] In the group that received a boost of 5xl0 10 vp of Ad26.ZIKV.00l on day 57, however, geometric mean MN50 titers increased almost 200-fold to 1980.3 on day 71 and remained at 1284 on day 85, demonstrating a strong boost effect.

[00192] In the group that received placebo after prime with lxl 0 11 vp of

Ad26.ZIKV.00l, geometric mean titers remained at 173.2 and 142.4 on day 71 and 85, respectively, while they substantially increased to 1016.8 and 1131 on days 71 and 85, respectively, in the group that received lxlO 11 vp of Ad26.ZIKV.00l as boost, again demonstrating a strong boost effect. All subjects receiving Ad26.ZIKV.00l at two doses, irrespective of the dose, developed MN50 titers >10 at both time points, resulting in a 100% seroconversion rate. In addition, 100% (14/14) in the group that received two vaccinations of Ad26.ZIKV.00l at 5x10 10 vp and 92.3% (12/13) in the group that received two vaccinations of Ad26.ZIKV.00l at lxlO 11 vp developed MN50 titers >100 - the threshold associated with protection against ZIKV challenge in animal models - at 28 days after the second vaccination. In contrast, in the groups that received placebo after the first vaccination of Ad26.ZIKV.00l at 5xl0 10 vp or at lxlO 11 vp the percentage of subjects whose MN50 titers reached this threshold was 20% (3/15) and 61.5% (8/13) (at 28 days after the second vaccination, respectively.

[00193] In summary, the results show that a robust humoral response was induced within 4 weeks after priming with Ad26. ZIKV.001, and that a second dose of

Ad26.ZIKV.00l administered eight weeks after priming was able to boost these responses to substantial levels. Table 4: Zika Microneutralization Assay (MN50): Actual Values, Fold Increases from Baseline and Percentage of Responders over Time; Per Protocol Immunogenicity Population (Study

VAC26911ZIK1001)

Ad26.ZIKV.001 Ad26.ZIKV.001 Ad26.ZIKV.001 Ad26.ZIKV.001

LD/LD LD/PL HD/HD HD/PL Placebo

ZIKV MN50 titer

Baseline

N 15 15 14 15 14

Geometric mean 5 (5;5) 5 (5;5) 5 (5;5) 5 (5;5) 5 (5;5)

(95% Cl)

MN50>=10 n (%) 0 0 0 0 0

MN50>=100 n (%) 0 0 0 0 0

Dav 15

N 15 15 14 15 14

Geometric mean 58.8 29.8 (9.6-92.6) 53.5 52.9 5 (5;5)

(95% Cl) (22.5;153.7) (17.1;167.2) (17.9;156.2)

Geometric mean fold 6.8 4.1 6.2 6.7 1 increase

Responders n (%) 12 (80.0%) 8 (53.3%) 11 (78.6%) 10 (66.7%) 0

MN50>=10 n (%) 12 (80.0%) 8 (53.3%) 11 (78.6%) 10 (66.7%) 0

MN50>= 100 n (%) 7 (46.7%) 4 (26.7%) 6 (42.9%) 7 (46.7%) 0

Day 29

N 15 15 14 14 13

Geometric mean 121.4 139.4 125 169 5 (5;5)

(95% Cl) (46.6;316.6) (50.1;388.3) (53.4;292.5) (63.4;450.7)

Geometric mean fold 12.7 15.3 13.1 17.8 1 increase

Responders n (%) 14 (93.3%) 13 (86.7%) 13 (92.9%) 13 (92.9%) 0

MN50>=10 n (%) 14 (93.3%) 13 (86.7%) 13 (92.9%) 13 (92.9%) 0

MN50>= 100 n (%) 8 (53.3%) 9 (60.0%) 8 (57.1%) 8 (57.1%) 0

Dav 57

N 14 15 13 13 13

Geometric mean 37.5 (14.7;95.8) 64.9 (32;131.6) 127 120.3 5 (5;5) (95% Cl) (49.1;328.3) (49.7;290.9)

Geometric mean fold 4.6 6.8 13.4 12 1 increase

Responders n (%) 10 (71.4%) 14 (93.3%) 12 (92.3%) 13 (100.0%) 0

MN50>=10 n (%) 10 (71.4%) 14 (93.3%) 12 (92.3%) 13 (100.0%) 0

MN50>= 100 n (%) 4 (28.6%) 5 (33.3%) 8 (61.5%) 8 (61.5%) 0

Dav 71

N 14 15 13 13 13

Geometric mean 1980.3 64.2 1016.8 173.2 5 (5;5)

(95% Cl) (1296;3025.7) (32.9; 125.1) (531.4;1945.6) (72.3;415.1)

Geometric mean fold 198 7 101.7 17.3 1 increase

Responders n (%) 14 (100.0%) 13 (86.7%) 13 (100.0%) 13 (100.0%) 0

MN50>=10 n (%) 14 (100.0%) 13 (86.7%) 13 (100.0%) 13 (100.0%) 0

MN50>=100 n (%) 14 (100.0%) 7 (46.7%) 12 (92.3%) 7 (53.8%) 0 Table 4: Zika Microneutralization Assay (MN50): Actual Values, Fold Increases from Baseline and Percentage of Responders over Time; Per Protocol Immunogenicity Population (Study

VAC26911ZIK1001)

Ad26.ZIKV.001 Ad26.ZIKV.001 Ad26.ZIKV.001 Ad26.ZIKV.001

LD/LD LD/PL HD/HD HD/PL Placebo

Day 85

N 14 15 13 13 13

Geometric mean 1284 44.8 (24.6;81.5) 1131 142.4 5 (5;5)

(95% Cl) (818.6;2014) (561.8;2276.6) (47.2;429.7)

Geometric mean fold 128.4 4.9 113.1 15 1 increase

Responders n (%) 14 (100.0%) 13 (86.7%) 13 (100.0%) 12 (92.3%) 0

MN50>=10 n (%) 14 (100.0%) 13 (86.7%) 13 (100.0%) 12 (92.3%) 0

MN50>=100 n (%) 14 (100.0%) 3 (20.0%) 12 (92.3%) 8 (61.5%) 0

N: number of subjects with data

Responder: 1) if baseline <LLOQ, R>=LLOQ 2) if baseline >=LLOQ, R=4-fold increase from baseline Fold Increase: 1) if baseline < LLOQ, FI=value post-baseline/LLOQ 2) if baseline >=LLOQ, FI=Value postbaseline/ Baseline Value

Note: Ad26.ZIKV.001 LD: Ad26.ZIKV.001 5xl0 10 vp; Ad26.ZIKV.001 HD: Ad26.ZIKV.001 lxlO 11 vp; PL: Placebo

[00194] It will be appreciated by those skilled in the art that changes could be made to the embodiments described above without departing from the broad inventive concept thereof. It is understood, therefore, that this invention is not limited to the particular embodiments disclosed, but it is intended to cover modifications within the spirit and scope of the present invention as defined by the present description.