Materials and methods for respiratory disease control in canines

Information

  • Patent Grant
  • 11865172
  • Patent Number
    11,865,172
  • Date Filed
    Friday, September 10, 2021
    2 years ago
  • Date Issued
    Tuesday, January 9, 2024
    4 months ago
Abstract
The subject invention pertains to isolated influenza virus that is capable of infecting canids and causing respiratory disease in the canid. The subject invention also pertains to compositions and methods for inducing an immune response against an influenza virus of the present invention. The subject invention also pertains to compositions and methods for identifying a virus of the invention and diagnosing infection of an animal with a virus of the invention.
Description
BACKGROUND OF THE INVENTION

“Kennel cough” or infectious tracheobronchitis (ITB) is an acute, contagious respiratory infection in dogs characterized mainly by coughing (Ford et al, 1998). Canine ITB is considered one of the most prevalent infectious respiratory diseases of dogs worldwide, and outbreaks can reach epidemic proportions when dogs are housed in high-density population environments such as kennels. Most outbreaks are due to direct dog-to-dog contact or aerosolization of respiratory secretions (Ford et al, 1998). The clinical signs are caused by infection with one or a combination of bacterial and viral agents that colonize the epithelium of the upper and lower respiratory tract. Canine parainfluenza virus (CPiV) and Bordetella bronchiseptica bacteria are the most common organisms isolated from affected dogs, but several other viruses such as canine distemper virus (CDV) and canine adenoviruses-1 and -2 (CAV-1, CAV-2), along with bacteria such as Streptococcus sp., Pasteurella multicoda and Escherichia coli, can influence the clinical course and outcome (Ford et al, 1998). While outbreaks occur most efficiently and rapidly in high-density populations with high morbidity, complicated respiratory infections and death are uncommon. Although life-threatening secondary bacterial pneumonia can develop, the majority of ITB cases are self-limiting and resolve without any treatment (Ford et al, 1998).


In July 1992, a respiratory infection presumed to be “kennel cough” became epidemic at several greyhound tracks in New England, Florida, West Virginia, Wisconsin, Kansas, Colorado, Oklahoma and Arizona. According to veterinarians, most of the affected dogs had a mild cough that resolved, but more than a dozen greyhounds developed an acute hemorrhagic pneumonia followed by rapid death (Greyhound Daily News, 1999).


In late 1998 to early 1999, several outbreaks of “kennel cough” occurred in racing greyhound kennels across the country, resulting in mandatory closure of tracks and quarantine of all racing greyhounds in the U.S. for several weeks (Greyhound Daily News, 1999). At one track in Florida (Palm Beach Kennel Club), coughing was recorded in nearly 40% of the dog population on a single day (Personal communication from Dr. William Duggar). Similar to the outbreak in 1992, the coughing resolved in most greyhounds, but 10 dogs in Florida died from a hemorrhagic pneumonia syndrome uncharacteristic of “kennel cough” (Putnam, 1999).


In March-April 2003, another outbreak of “kennel cough” occurred at greyhound tracks in the eastern U.S. The outbreak is believed to have originated in kennels at four tracks in Florida and caused the suspension of racing and quarantine of dogs for almost three weeks. Nearly 25% of the dogs at the track in West Palm Beach were affected, while almost 50% of the 1400 dogs at Derby Lane in St. Petersburg developed coughing. Again, most dogs recovered, but several dogs have died from the respiratory infection. The estimated economic impact of the respiratory outbreak at the Derby Lane track alone was $2 million.


There are no published reports documenting the etiology or clinicopathology of the “kennel cough” epidemics in racing greyhound kennels in 1992, 1998-1999, or 2003. The assumption has been that the infections were due to CPiV and/or B. bronchiseptica, the two most common causes of kennel cough. Unsubstantiated communications such as web sites have attributed the fatal hemorrhagic pneumonias reported in some of the coughing dogs to infection with β-hemolytic Streptococcus equi subspecies zooepidemicus, and refer to the syndrome as “canine streptococcal toxic shock.”


Transmission of virus from one host species to another is a crucial feature of the ecology and epidemiology of influenza virus (Webster, 1998). Two basic mechanisms of interspecies transmission of influenza virus are possible (Webster et al., 1992; Lipatov et al., 2004). One is the direct transfer of an essentially unaltered virus from one species to another. Examples of this mechanism include the recent human infections with the H5N1 subtype of avian influenza virus (Subbarao et al., 1998; Peiris et al., 2004; Guan et al., 2004) and possibly the pandemic of 1918, known as Spanish flu (Reid et al., 2004). The second mechanism is a consequence of the segmented nature of the influenza genome. Co-infection of a host with viruses from different species can result in reassortment of the segmented viral genes and the generation of a recombinant with the ability to infect other species. For example, novel viruses generated by gene reassortment between avian and human influenza viruses resulted in human influenza pandemics in 1957 and 1968 (Webster et al., 1992; Lipatov et al., 2004; Kawaoka et al., 1989).


Most direct transmissions of unaltered influenza viruses from the natural host species to a different species are terminal events because sustained transmission between individuals of the new species fails to occur. Multiple virus-host interactions are necessary for replication and horizontal transmission and provide a formidable barrier to perpetuation of influenza viruses in the new host (Webby et al., 2004). Therefore, establishment of new host-specific lineages of influenza virus is uncommon and has only occurred in domestic poultry, pigs, horses, and humans (Webster et al., 1992; Lipatov et al., 2004).


Because of the serious nature of influenza virus infection, there remains a need for methods for diagnosing, preventing, and treating infection by influenza virus.


BRIEF SUMMARY OF THE INVENTION

The subject invention pertains to isolated influenza virus that is capable of infecting canids and causing respiratory disease in the canid. The subject invention also pertains to compositions and methods for inducing an immune response against an influenza virus of the present invention. The subject invention also pertains to compositions and methods for identifying a virus of the invention and diagnosing infection of an animal with a virus of the invention.


One aspect of the invention relates to vaccines and methods for protecting canines from canine influenza, kits comprising such vaccines, and methods for using such vaccines. This protection includes preventing, reducing the risk of, delaying the onset of, reducing the spread of, ameliorating, suppressing, and/or eradicating the influenza and/or one or more (typically two or more) of its symptoms. It is believed that the vaccines, kits, and methods of this invention are generally suitable for use with canines. Canines include wild, zoo, and domestic canines, such as wolves, coyotes, and foxes. Canines also include dogs, particularly domestic dogs, such as, for example, pure-bred and/or mongrel companion dogs, show dogs, working dogs, herding dogs, hunting dogs, guard dogs, police dogs, racing dogs, and/or laboratory dogs.


This invention is also directed, in part, to a method for protecting a canine from an influenza virus infection (i.e., preventing, reducing the risk of, delaying the onset of, suppressing, ameliorating, or eradicating an influenza virus infection). The method comprises administering a therapeutically effective amount of a vaccine that comprises at least one equine influenza virus antigen, at least one H3 influenza virus antigen, and/or at least one H7 influenza virus antigen.


This invention also is directed, in part, to a method for protecting a canine from respiratory lesions (i.e., preventing, reducing the risk of, delaying the onset of, suppressing, ameliorating, or eradicating respiratory lesions) caused by canine influenza virus. The method comprises administering to the canine a therapeutically effective amount of a vaccine that comprises at least one equine influenza virus antigen, at least one H3 influenza virus antigen, and/or at least one H7 influenza virus antigen.


This invention also is directed, in part, to a method for protecting a canine from having canine influenza virus in nasal or oral secretion (i.e., preventing, reducing the risk of, delaying the onset of, suppressing, ameliorating, or eradicating canine influenza virus in nasal or oral secretion) caused by canine influenza virus infection. The method comprises administering to the canine a therapeutically effective amount of a vaccine that comprises at least one equine influenza virus antigen, at least one H3 influenza virus antigen, and/or at least one H7 influenza virus antigen.


This invention also is directed, in part, to a canine influenza vaccine. In some embodiments, for example, the vaccine comprises a therapeutically effective amount of at least one equine influenza virus antigen, at least one H3 influenza virus antigen, and/or at least one H7 influenza virus antigen.


This invention also is directed, in part, to a kit for protecting a canine from influenza virus infection. The kit comprises a therapeutically effective amount of a vaccine that comprises at least one equine influenza virus antigen, at least one H3 influenza virus antigen, and/or at least one H7 influenza virus antigen. In addition, the kit comprises at least one of the following:

    • an apparatus for administering the vaccine to the canine,
    • a pharmaceutically acceptable excipient that aids in administering the vaccine to the canine,
    • a pharmaceutically acceptable excipient that enhances the canine's immune response to the vaccine,
    • a food to be consumed by the canine simultaneously with the vaccine, and/or
    • a treat to be consumed by the canine simultaneously with the vaccine.


Further benefits of Applicants' invention will be apparent to one skilled in the art from reading this specification.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1B show phylogenetic relationships among the hemagglutinin genes. FIG. 1A shows a tree of HA genes from representative canine, human, avian, swine, and equine isolates, including A/Budgerigar/Hokkaido/1/77 (H4) as outgroup. FIG. 1B shows a tree of the canine influenza virus HA genes with contemporary and older equine HA genes, using A/Duck/Ukraine/63 (H3) as outgroup. Phylogenetic trees were inferred from nucleotide sequences by the neighbor joining method and bootstrap analysis values ≥90% are shown. The bar denotes the number of nucleotide changes per unit length of the horizontal tree branches.



FIGS. 2A-2B show immunohistochemical detection of influenza H3 antigen in the lungs. Lung tissue sections were probed with a mouse monoclonal antibody to H3 hemagglutinin and binding was detected by immunoperoxidase reaction (brown precipitate). FIG. 2A shows bronchial epithelium from a greyhound with spontaneous disease. Viral H3 antigen was detected in bronchial epithelial cell cytoplasm and in macrophages in airway lumens and in alveolar spaces. FIG. 2B shows bronchial epithelium from a dog 5 days after inoculation with A/canine/Florida/43/2004 (H3N8). Viral H3 antigen was detected in bronchial epithelial cell cytoplasm. Scale bar, 66 μm.



FIGS. 3A and 3B show the characteristic histological changes in the bronchi of greyhounds that died from hemorrhagic pneumonia associated with influenza virus infection. The tissues are stained with H&E. FIG. 3A: Normal bronchus with ciliated epithelial cells, mucous cells, and basal cells. FIG. 3B: Bronchus from a greyhound with spontaneous influenza. There is necrosis and erosion of the bronchial ciliated epithelial cells. Scale bar, 100 μm.



FIGS. 4A-4B shows phylogenetic relationships among the H3 hemagglutinin genes. FIG. 4A shows a phylogenetic tree of the canine influenza virus HA genes with contemporary and older equine HA genes. FIG. 4B shows a phylogenetic tree of the canine influenza virus HA protein with contemporary and older equine HA. Phylogenetic trees were inferred from genetic or amino acid sequences by the neighbor joining method and bootstrap analysis values ≥80% are shown. The bar denotes the number of amino acid changes per unit length of the horizontal tree branches.



FIG. 5 shows Influenza virus H3 protein in epithelial cells of bronchi and bronchial glands in lungs of dogs that died of pneumonia associated with influenza virus infection. Upper panels: Erosion of ciliated bronchial epithelial cells in bronchi. Tissues were stained with H&E. Lower panels: Influenza virus H3 protein in the cytoplasm of bronchial (left) and bronchial gland (right) epithelial cells. Tissues were stained with a monoclonal antibody to influenza H3 detected by immunoperoxidase reaction (brown precipitate) and counterstained with hematoxylin.



FIGS. 6A-6D show amplification plots of H3 and Matrix genes (FIG. 6A and FIG. 6B) obtained from the amplification of 10-fold serially diluted in vitro transcribed RNA standards. Standard curves of H3 and Matrix genes (FIG. 6C and FIG. 6D) constructed by plotting the log of starting RNA concentrations against the threshold cycle (Ct) obtained from each dilution.



FIG. 7 shows sensitivity of Directigen Flu A was tested with 10-fold serially diluted virus stocks including A/Wyoming/3/2003 and A/canine/FL/242/2003. The purple triangle indicates positive result.





BRIEF DESCRIPTION OF THE SEQUENCES

SEQ ID NO: 1 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding a PB2 protein that can be used according to the present invention.


SEQ ID NO: 2 is the amino acid sequence encoded by SEQ ID NO: 1.


SEQ ID NO: 3 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding a PB1 protein that can be used according to the present invention.


SEQ ID NO: 4 is the amino acid sequence encoded by SEQ ID NO: 3.


SEQ ID NO: 5 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding a PA protein that can be used according to the present invention.


SEQ ID NO: 6 is the amino acid sequence encoded by SEQ ID NO: 5.


SEQ ID NO: 7 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding an NS protein that can be used according to the present invention.


SEQ ID NO: 8 is the amino acid sequence encoded by SEQ ID NO: 7.


SEQ ID NO: 9 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding an NP protein that can be used according to the present invention.


SEQ ID NO: 10 is the amino acid sequence encoded by SEQ ID NO: 9.


SEQ ID NO: 11 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding an NA protein that can be used according to the present invention.


SEQ ID NO: 12 is the amino acid sequence encoded by SEQ ID NO: 11.


SEQ ID NO: 13 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding an MA protein that can be used according to the present invention.


SEQ ID NO: 14 is the amino acid sequence encoded by SEQ ID NO: 13.


SEQ ID NO: 15 is a nucleotide sequence of a canine influenza virus (Florida/43/04) encoding an HA protein that can be used according to the present invention.


SEQ ID NO: 16 is the amino acid sequence encoded by SEQ ID NO: 15.


SEQ ID NO: 17 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding a PB2 protein that can be used according to the present invention.


SEQ ID NO: 18 is the amino acid sequence encoded by SEQ ID NO: 17.


SEQ ID NO: 19 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding a PB1 protein that can be used according to the present invention.


SEQ ID NO: 20 is the amino acid sequence encoded by SEQ ID NO: 19.


SEQ ID NO: 21 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding a PA protein that can be used according to the present invention.


SEQ ID NO: 22 is the amino acid sequence encoded by SEQ ID NO: 21.


SEQ ID NO: 23 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding an NS protein that can be used according to the present invention.


SEQ ID NO: 24 is the amino acid sequence encoded by SEQ ID NO: 23.


SEQ ID NO: 25 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding an NP protein that can be used according to the present invention.


SEQ ID NO: 26 is the amino acid sequence encoded by SEQ ID NO: 25.


SEQ ID NO: 27 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding an NA protein that can be used according to the present invention.


SEQ ID NO: 28 is the amino acid sequence encoded by SEQ ID NO: 27.


SEQ ID NO: 29 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding an MA protein that can be used according to the present invention.


SEQ ID NO: 30 is the amino acid sequence encoded by SEQ ID NO: 29.


SEQ ID NO: 31 is a nucleotide sequence of a canine influenza virus (FL/242/03) encoding an HA protein that can be used according to the present invention.


SEQ ID NO: 32 is the amino acid sequence encoded by SEQ ID NO: 31.


SEQ ID NO: 33 is the mature form of the HA protein shown in SEQ ID NO: 16 wherein the N-terminal 16 amino acid signal sequence has been removed.


SEQ ID NO: 34 is the mature form of the HA protein shown in SEQ ID NO: 32 wherein the N-terminal 16 amino acid signal sequence has been removed.


SEQ ID NO: 35 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 36 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 37 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 38 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 39 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 41 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 42 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 43 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 44 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 45 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 46 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 47 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding a PB2 protein that can be used according to the present invention.


SEQ ID NO: 48 is the amino acid sequence encoded by SEQ ID NO: 47.


SEQ ID NO: 49 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding a PB1 protein that can be used according to the present invention.


SEQ ID NO: 50 is the amino acid sequence encoded by SEQ ID NO: 49.


SEQ ID NO: 51 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding a PA protein that can be used according to the present invention.


SEQ ID NO: 52 is the amino acid sequence encoded by SEQ ID NO: 51.


SEQ ID NO: 53 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding an NS protein that can be used according to the present invention.


SEQ ID NO: 54 is the amino acid sequence encoded by SEQ ID NO: 53.


SEQ ID NO: 55 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding an NP protein that can be used according to the present invention.


SEQ ID NO: 56 is the amino acid sequence encoded by SEQ ID NO: 55.


SEQ ID NO: 57 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding an NA protein that can be used according to the present invention.


SEQ ID NO: 58 is the amino acid sequence encoded by SEQ ID NO: 57.


SEQ ID NO: 59 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding an MA protein that can be used according to the present invention.


SEQ ID NO: 60 is the amino acid sequence encoded by SEQ ID NO: 59.


SEQ ID NO: 61 is a nucleotide sequence of a canine influenza virus (Miami/2005) encoding an HA protein that can be used according to the present invention.


SEQ ID NO: 62 is the amino acid sequence encoded by SEQ ID NO: 61.


SEQ ID NO: 63 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding a PB2 protein that can be used according to the present invention.


SEQ ID NO: 64 is the amino acid sequence encoded by SEQ ID NO: 63.


SEQ ID NO: 65 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding a PB1 protein that can be used according to the present invention.


SEQ ID NO: 66 is the amino acid sequence encoded by SEQ ID NO: 65.


SEQ ID NO: 67 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding a PA protein that can be used according to the present invention.


SEQ ID NO: 68 is the amino acid sequence encoded by SEQ ID NO: 67.


SEQ ID NO: 69 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding an NS protein that can be used according to the present invention.


SEQ ID NO: 70 is the amino acid sequence encoded by SEQ ID NO: 69.


SEQ ID NO: 71 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding an NP protein that can be used according to the present invention.


SEQ ID NO: 72 is the amino acid sequence encoded by SEQ ID NO: 71.


SEQ ID NO: 73 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding an NA protein that can be used according to the present invention.


SEQ ID NO: 74 is the amino acid sequence encoded by SEQ ID NO: 73.


SEQ ID NO: 75 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding an MA protein that can be used according to the present invention.


SEQ ID NO: 76 is the amino acid sequence encoded by SEQ ID NO: 75.


SEQ ID NO: 77 is a nucleotide sequence of a canine influenza virus (Jacksonville/2005) encoding an HA protein that can be used according to the present invention.


SEQ ID NO: 78 is the amino acid sequence encoded by SEQ ID NO: 77.


SEQ ID NO: 79 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 80 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 81 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 82 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 83 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 84 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 85 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 86 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 87 is an oligonucleotide that can be used according to the present invention.


SEQ ID NO: 88 is an oligonucleotide that can be used according to the present invention.


DETAILED DISCLOSURE OF THE INVENTION

The subject invention concerns isolated influenza virus that is capable of infecting canids and causing respiratory disease. In one embodiment, an influenza virus of the invention comprises a polynucleotide which encodes a protein having an amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a specific embodiment, the polynucleotide comprises the nucleotide sequence shown in any of SEQ ID Nos: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, or a fragment or variant thereof. Influenza virus of the present invention can have an HA subtype of H1, H2, H3, H4, H5, H6, H7, H8, and H9, H10, H11, H12, H13, H14, H15, or H16 or an NA subtype of N1, N2, N3, N4, N5, N6, N7, N8, OR N9. In a specific embodiment, an influenza virus of the present invention is a subtype H3. Virus can be isolated from infected dogs and cultured in cells or eggs according to methods described herein. In an exemplified embodiment, the influenza virus is an influenza A virus.


The subject invention also concerns polynucleotides that comprise all or part of a gene or genes or a genomic segment of an influenza virus of the present invention. In one embodiment, a polynucleotide of the invention comprises an influenza hemagglutinin (HA) gene, neuraminidase (NA) gene, nucleoprotein (NP) gene, matrix protein (MA or M) gene, polymerase basic (PB) protein gene, polymerase acidic (PA) protein gene, non-structural (NS) protein gene, or a functional fragment or variant of any of these genes. In a specific embodiment, a polynucleotide of the invention comprises the hemagglutinin (HA) gene, or a functional fragment or variant thereof. In a further embodiment, the HA gene encodes a hemagglutinin protein having one or more of the following: a serine at position 83; a leucine at position 222; a threonine at position 328; and/or a threonine at position 483, versus the amino acid sequence of equine H3 consensus sequence. In one embodiment, the HA gene encodes a polypeptide having an amino acid sequence shown in SEQ ID NOs: 16, 32, 62, or 78, or a functional and/or immunogenic fragment or variant thereof. In a specific embodiment, the HA gene comprises a nucleotide sequence shown in SEQ ID NOs: 15, 31, 61, or 77.


In one embodiment, a polynucleotide of the invention encodes a polypeptide having the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a specific embodiment, the polynucleotide encoding the amino acid sequence shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, comprises the nucleotide sequence shown in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, respectively, or a sequence encoding a functional and/or immunogenic fragment or variant of any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78. Thus, the subject invention concerns polynucleotide sequences comprising the nucleotide sequence shown in any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, or a fragment or variant, including a degenerate variant, of any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77. In a further specific embodiment, a polynucleotide of the invention can comprise: Nucleotides 1-2271 of SEQ ID NO: 3; Nucleotides 1-2148 of SEQ ID NO: 5; Nucleotides 1-657 of SEQ ID NO: 7; Nucleotides 1-1494 of SEQ ID NO: 9; Nucleotides 1-1410 of SEQ ID NO: 11; Nucleotides 1-756 of SEQ ID NO: 13; Nucleotides 1-1695 of SEQ ID NO: 15; Nucleotides 1-2271 of SEQ ID NO: 19; Nucleotides 1-2148 of SEQ ID NO: 21; Nucleotides 1-657 of SEQ ID NO: 23; Nucleotides 1-1494 of SEQ ID NO: 25; Nucleotides 1-756 of SEQ ID NO: 29; Nucleotides 1-1695 of SEQ ID NO: 31; Nucleotides 1-2277 of SEQ ID NO: 47; Nucleotides 1-2271 of SEQ ID NO: 49; Nucleotides 1-2148 of SEQ ID NO: 51; Nucleotides 1-690 of SEQ ID NO: 53; Nucleotides 1-1494 of SEQ ID NO: 55; Nucleotides 1-1410 of SEQ ID NO: 57; Nucleotides 1-756 of SEQ ID NO: 59; Nucleotides 1-1695 of SEQ ID NO: 61; Nucleotides 1-2277 of SEQ ID NO: 63; Nucleotides 1-2271 of SEQ ID NO: 65; Nucleotides 1-2148 of SEQ ID NO: 67; Nucleotides 1-690 of SEQ ID NO: 69; Nucleotides 1-1494 of SEQ ID NO: 71; Nucleotides 1-1410 of SEQ ID NO: 73; Nucleotides 1-756 of SEQ ID NO: 75; and Nucleotides 1-1695 of SEQ ID NO: 77. Nucleotide and amino acid sequences of viral polynucleotide and polypeptide sequences contemplated within the scope of the present invention have also been deposited with GenBank at accession Nos. DQ124147 through DQ124161 and DQ124190, the disclosure of which is incorporated herein by reference.


The subject invention also concerns polypeptides encoded by polynucleotides of an influenza virus of the present invention. The subject invention also concerns functional and/or immunogenic fragments and variants of the subject polypeptides. Polypeptides contemplated include HA protein, NA protein, NS protein, nucleoprotein, polymerase basic protein, polymerase acidic protein, and matrix protein of an influenza virus of the invention. In an exemplified embodiment, a polypeptide of the invention has an amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof.


The subject invention also concerns polynucleotide expression constructs comprising a polynucleotide sequence of the present invention. In one embodiment, an expression construct of the invention comprises a polynucleotide sequence encoding a polypeptide comprising an amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a specific embodiment, the polynucleotide encoding the amino acid sequence shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78 comprises the nucleotide sequence shown in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, respectively, or a sequence encoding a functional and/or immunogenic fragment or variant of any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78. Thus, the subject invention concerns expression constructs comprising a polynucleotide sequence comprising the nucleotide sequence shown in any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, or a fragment or variant, including a degenerate variant, of any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77. In a preferred embodiment, an expression construct of the present invention provides for overexpression of an operably linked polynucleotide of the invention.


Expression constructs of the invention generally include regulatory elements that are functional in the intended host cell in which the expression construct is to be expressed. Thus, a person of ordinary skill in the art can select regulatory elements for use in, for example, human host cells, mammalian host cells, insect host cells, yeast host cells, bacterial host cells, and plant host cells. In one embodiment, the regulatory elements are ones that are functional in canine cells. Regulatory elements include promoters, transcription termination sequences, translation termination sequences, enhancers, and polyadenylation elements. As used herein, the term “expression construct” refers to a combination of nucleic acid sequences that provides for transcription of an operably linked nucleic acid sequence. As used herein, the term “operably linked” refers to a juxtaposition of the components described wherein the components are in a relationship that permits them to function in their intended manner. In general, operably linked components are in contiguous relation.


An expression construct of the invention can comprise a promoter sequence operably linked to a polynucleotide sequence encoding a polypeptide of the invention. Promoters can be incorporated into a polynucleotide using standard techniques known in the art. Multiple copies of promoters or multiple promoters can be used in an expression construct of the invention. In a preferred embodiment, a promoter can be positioned about the same distance from the transcription start site in the expression construct as it is from the transcription start site in its natural genetic environment. Some variation in this distance is permitted without substantial decrease in promoter activity. A transcription start site is typically included in the expression construct. Preferably, the promoter associated with an expression construct of the invention provides for overexpression of an operably linked polynucleotide of the invention.


Promoters for use with an expression construct of the invention in eukaryotic cells can be of viral or cellular origin. Viral promoters include, but are not limited to, cytomegalovirus (CMV) gene promoters, SV40 early or late promoters, or Rous sarcoma virus (RSV) gene promoters. Promoters of cellular origin include, but are not limited to, desmin gene promoter and actin gene promoter Promoters suitable for use with an expression construct of the invention in yeast cells include, but are not limited to, 3-phosphoglycerate kinase promoter, glyceraldehyde-3-phosphate dehydrogenase promoter, metallothionein promoter, alcohol dehydrogenase-2 promoter, and hexokinase promoter.


If the expression construct is to be provided in or introduced into a plant cell, then plant viral promoters, such as, for example, a cauliflower mosaic virus (CaMV) 35S (including the enhanced CaMV 35S promoter (see, for example U.S. Pat. No. 5,106,739 and An, 1997)) or a CaMV 19S promoter can be used. Other promoters that can be used for expression constructs in plants include, for example, prolifera promoter, Ap3 promoter, heat shock promoters, T-DNA 1′- or 2′-promoter of A. tumefaciens, polygalacturonase promoter, chalcone synthase A (CHS-A) promoter from petunia, tobacco PR-la promoter, ubiquitin promoter, actin promoter, alcA gene promoter, pin2 promoter (Xu et al., 1993), maize WipI promoter, maize trpA gene promoter (U.S. Pat. No. 5,625,136), maize CDPK gene promoter, and RUBISCO SSU promoter (U.S. Pat. No. 5,034,322) can also be used. Root-specific promoters, such as any of the promoter sequences described in U.S. Pat. No. 6,455,760 or U.S. Pat. No. 6,696,623, or in published U.S. patent application Nos. 20040078841; 20040067506; 20040019934; 20030177536; 20030084486; or 20040123349, can be used with an expression construct of the invention. Constitutive promoters (such as the CaMV, ubiquitin, actin, or NOS promoter), develoμmentally-regulated promoters, and inducible promoters (such as those promoters than can be induced by heat, light, hormones, or chemicals) are also contemplated for use with polynucleotide expression constructs of the invention. Tissue-specific promoters, for example fruit-specific promoters, such as the E8 promoter of tomato (accession number: AF515784; Good et al. (1994)) can also be used. Seed-specific promoters such as the promoter from a β-phaseolin gene (for example, of kidney bean) or a glycinin gene (for example, of soybean), and others, can also be used.


For expression in prokaryotic systems, an expression construct of the invention can comprise promoters such as, for example, alkaline phosphatase promoter, tryptophan (trp) promoter, lambda PL promoter, β-lactamase promoter, lactose promoter, phoA promoter, T3 promoter, T7 promoter, or tac promoter (de Boer et al., 1983).


Expression constructs of the invention may optionally contain a transcription termination sequence, a translation termination sequence, a sequence encoding a signal peptide, and/or enhancer elements. Transcription termination regions can typically be obtained from the 3′ untranslated region of a eukaryotic or viral gene sequence. Transcription termination sequences can be positioned downstream of a coding sequence to provide for efficient termination. A signal peptide sequence is a short amino acid sequence typically present at the amino terminus of a protein that is responsible for the relocation of an operably linked mature polypeptide to a wide range of post-translational cellular destinations, ranging from a specific organelle compartment to sites of protein action and the extracellular environment. Targeting gene products to an intended cellular and/or extracellular destination through the use of an operably linked signal peptide sequence is contemplated for use with the polypeptides of the invention. Classical enhancers are cis-acting elements that increase gene transcription and can also be included in the expression construct. Classical enhancer elements are known in the art, and include, but are not limited to, the CaMV 35S enhancer element, cytomegalovirus (CMV) early promoter enhancer element, and the SV40 enhancer element. Intron-mediated enhancer elements that enhance gene expression are also known in the art. These elements must be present within the transcribed region and are orientation dependent.


DNA sequences which direct polyadenylation of mRNA transcribed from the expression construct can also be included in the expression construct, and include, but are not limited to, an octopine synthase or nopaline synthase signal.


Expression constructs can also include one or more dominant selectable marker genes, including, for example, genes encoding antibiotic resistance and/or herbicide-resistance for selecting transformed cells. Antibiotic-resistance genes can provide for resistance to one or more of the following antibiotics: hygromycin, kanamycin, bleomycin, G418, streptomycin, paromomycin, neomycin, and spectinomycin. Kanamycin resistance can be provided by neomycin phosphotransferase (NPT II). Herbicide-resistance genes can provide for resistance to phosphinothricin acetyltransferase or glyphosate. Other markers used for cell transformation screening include, but are not limited to, genes encoding β-glucuronidase (GUS), β-galactosidase, luciferase, nopaline synthase, chloramphenicol acetyltransferase (CAT), green fluorescence protein (GFP), or enhanced GFP (Yang et al., 1996).


The subject invention also concerns polynucleotide vectors comprising a polynucleotide sequence of the invention that encodes a polypeptide of the invention. Unique restriction enzyme sites can be included at the 5′ and 3′ ends of an expression construct or polynucleotide of the invention to allow for insertion into a polynucleotide vector. As used herein, the term “vector” refers to any genetic element, including for example, plasmids, cosmids, chromosomes, phage, virus, and the like, which is capable of replication when associated with proper control elements and which can transfer polynucleotide sequences between cells. Vectors contain a nucleotide sequence that permits the vector to replicate in a selected host cell. A number of vectors are available for expression and/or cloning, and include, but are not limited to, pBR322, pUC series, M13 series, pGEM series, and pBLUESCRIPT vectors (Stratagene, La Jolla, CA and Promega, Madison, WI).


The subject invention also concerns oligonucleotide probes and primers, such as polymerase chain reaction (PCR) primers, that can hybridize to a coding or non-coding sequence of a polynucleotide of the present invention. Oligonucleotide probes of the invention can be used in methods for detecting influenza virus nucleic acid sequences. Oligonucleotide primers of the invention can be used in PCR methods and other methods involving nucleic acid amplification. In a preferred embodiment, a probe or primer of the invention can hybridize to a polynucleotide of the invention under stringent conditions. Probes and primers of the invention can optionally comprise a detectable label or reporter molecule, such as fluorescent molecules, enzymes, radioactive moiety, and the like. Probes and primers of the invention can be of any suitable length for the method or assay in which they are being employed. Typically, probes and primers of the invention will be 10 to 500 or more nucleotides in length. Probes and primers that are 10 to 20, 21 to 30, 31 to 40, 41 to 50, 51 to 60, 61 to 70, 71 to 80, 81 to 90, 91 to 100, or 101 or more nucleotides in length are contemplated within the scope of the invention. In one embodiment, probes and primers are any of 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides in length. Probes and primers of the invention can have complete (100%) nucleotide sequence identity with the polynucleotide sequence, or the sequence identity can be less than 100%. For example, sequence identity between a probe or primer and a sequence can be 99%, 98%, 97%, 96%, 95%, 90%, 85%, 80%, 75%, 70% or any other percentage sequence identity so long as the probe or primer can hybridize under stringent conditions to a nucleotide sequence of a polynucleotide of the invention. Exemplified probes and primers of the invention include those having the nucleotide sequence shown in any of SEQ ID NO: 35, SEQ ID NO: 36, SEQ ID NO: 37, SEQ ID NO: 38, SEQ ID NO: 39, SEQ ID NO: 40, SEQ ID NO: 41, SEQ ID NO: 42, SEQ ID NO: 43, SEQ ID NO: 44, SEQ ID NO: 45, and SEQ ID NO: 46, or a functional fragment or variant of any of the SEQ ID NOs: 35-46.


As used herein, the terms “nucleic acid,” “polynucleotide,” and “oligonucleotide” refer to a deoxyribonucleotide, ribonucleotide, or a mixed deoxyribonucleotide and ribonucleotide polymer in either single- or double-stranded form, and unless otherwise limited, would encompass known analogs of natural nucleotides that can function in a similar manner as naturally-occurring nucleotides. Polynucleotide sequences include the DNA strand sequence that can be transcribed into RNA and the RNA strand that can be translated into protein. The complementary sequence of any nucleic acid, polynucleotide, or oligonucleotide of the present invention is also contemplated within the scope of the invention. Polynucleotide sequences also include both full-length sequences as well as shorter sequences derived from the full-length sequences. The subject invention also encompasses those polynucleotides that are complementary in sequence to the polynucleotides disclosed herein. Polynucleotides and polypeptides of the invention can be provided in purified or isolated form.


Because of the degeneracy of the genetic code, a variety of different polynucleotide sequences can encode a polypeptide of the present invention. A table showing all possible triplet codons (and where U also stands for T) and the amino acid encoded by each codon is described in Lewin (1985). In addition, it is well within the skill of a person trained in the art to create alternative polynucleotide sequences encoding the same, or essentially the same, polypeptides of the subject invention. These degenerate variant and alternative polynucleotide sequences are within the scope of the subject invention. As used herein, references to “essentially the same” sequence refers to sequences which encode amino acid substitutions, deletions, additions, or insertions which do not materially alter the functional and/or immunogenic activity of the polypeptide encoded by the polynucleotides of the present invention.


The subject invention also concerns variants of the polynucleotides of the present invention that encode polypeptides of the invention. Variant sequences include those sequences wherein one or more nucleotides of the sequence have been substituted, deleted, and/or inserted. The nucleotides that can be substituted for natural nucleotides of DNA have a base moiety that can include, but is not limited to, inosine, 5-fluorouracil, 5-bromouracil, hypoxanthine, 1-methylguanine, 5-methylcytosine, and tritylated bases. The sugar moiety of the nucleotide in a sequence can also be modified and includes, but is not limited to, arabinose, xylulose, and hexose. In addition, the adenine, cytosine, guanine, thymine, and uracil bases of the nucleotides can be modified with acetyl, methyl, and/or thio groups. Sequences containing nucleotide substitutions, deletions, and/or insertions can be prepared and tested using standard techniques known in the art.


Substitution of amino acids other than those specifically exemplified or naturally present in a polypeptide of the invention are also contemplated within the scope of the present invention. For example, non-natural amino acids can be substituted for the amino acids of a polypeptide, so long as the polypeptide having the substituted amino acids retains substantially the same functional activity as the polypeptide in which amino acids have not been substituted. Examples of non-natural amino acids include, but are not limited to, ornithine, citrulline, hydroxyproline, homoserine, phenylglycine, taurine, iodotyrosine, 2,4-diaminobutyric acid, α-amino isobutyric acid, 4-aminobutyric acid, 2-amino butyric acid, γ-amino butyric acid, ε-amino hexanoic acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3-amino propionic acid, norleucine, norvaline, sarcosine, homocitrulline, cysteic acid, τ-butylglycine, τ-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, C-methyl amino acids, N-methyl amino acids, and amino acid analogues in general. Non-natural amino acids also include amino acids having derivatized side groups. Furthermore, any of the amino acids in the protein can be of the D (dextrorotary) form or L (levorotary) form. Allelic variants of a protein sequence of a polypeptide of the present invention are also encompassed within the scope of the invention.


Amino acids can be generally categorized in the following classes: non-polar, uncharged polar, basic, and acidic. Conservative substitutions whereby a polypeptide of the present invention having an amino acid of one class is replaced with another amino acid of the same class fall within the scope of the subject invention so long as the polypeptide having the substitution still retains substantially the same functional activity as the polypeptide that does not have the substitution. Polynucleotides encoding a polypeptide having one or more amino acid substitutions in the sequence are contemplated within the scope of the present invention. Table 11 below provides a listing of examples of amino acids belonging to each class. Single letter amino acid abbreviations are defined in Table 12.


Fragments and variants of polypeptides of influenza virus of the present invention can be generated using standard methods known in the art and tested for the presence of function or immunogenecity using standard techniques known in the art. For example, for testing fragments and/or variants of a neuraminidase polypeptide of the invention, enzymatic activity can be assayed. Thus, an ordinarily skilled artisan can readily prepare and test fragments and variants of a polypeptide of the invention and determine whether the fragment or variant retains activity relative to full-length or a non-variant polypeptide.


Polynucleotides and polypeptides contemplated within the scope of the subject invention can also be defined in terms of more particular identity and/or similarity ranges with those sequences of the invention specifically exemplified herein. The sequence identity will typically be greater than 60%, preferably greater than 75%, more preferably greater than 80%, even more preferably greater than 90%, and can be greater than 95%. The identity and/or similarity of a sequence can be 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, or 99% as compared to a sequence exemplified herein. Unless otherwise specified, as used herein percent sequence identity and/or similarity of two sequences can be determined using the algorithm of Karlin and Altschul (1990), modified as in Karlin and Altschul (1993). Such an algorithm is incorporated into the NBLAST and XBLAST programs of Altschul et al. (1990). BLAST searches can be performed with the NBLAST program, score=100, wordlength=12, to obtain sequences with the desired percent sequence identity. To obtain gapped alignments for comparison purposes, Gapped BLAST can be used as described in Altschul et al. (1997). When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (NBLAST and)(BLAST) can be used. See NCBI/NIH website.


The subject invention also contemplates those polynucleotide molecules having sequences which are sufficiently homologous with the polynucleotide sequences exemplified herein so as to permit hybridization with that sequence under standard stringent conditions and standard methods (Maniatis et al., 1982). As used herein, “stringent” conditions for hybridization refers to conditions wherein hybridization is typically carried out overnight at 20-25 C below the melting temperature (Tm) of the DNA hybrid in 6×SSPE, 5×Denhardt's solution, 0.1% SDS, 0.1 mg/ml denatured DNA. The melting temperature, Tm, is described by the following formula (Beltz et al., 1983):


Tm=81.5 C+16.6 Log[Na+]+0.41(% G+C)−0.61(% formamide)−600/length of duplex in base pairs.


Washes are typically carried out as follows:

    • (1) Twice at room temperature for 15 minutes in 1×SSPE, 0.1% SDS (low stringency wash).
    • (2) Once at Tm-20 C for 15 minutes in 0.2× SSPE, 0.1% SDS (moderate stringency wash).


The subject invention also concerns viral proteins and peptides encoded by the genes of an influenza virus of the present invention. In one embodiment, the viral protein is a mature HA protein. In a specific embodiment, the mature HA protein comprises one or more of the following: a serine at position 82; a leucine at position 221; a threonine at position 327; and/or a threonine at position 482. In an exemplified embodiment, the mature HA protein has an amino acid sequence shown in SEQ ID NO: 33 or SEQ ID NO: 34, or a functional and/or immunogenic fragment or variant of SEQ ID NO: 33 or SEQ ID NO: 34. In another embodiment, the viral protein is an NA protein, NS protein, PB protein, PA protein, or MA protein. Viral proteins and peptides of the invention can be used to generate antibodies that bind specifically to the protein or peptide. Viral proteins and peptides of the present invention can also be used as immunogens and in vaccine compositions.


The subject invention also concerns compositions and methods for inducing an immune response against an influenza virus that is capable of infecting a susceptible host animal and causing respiratory disease. The invention can be used to induce an immune response against an influenza virus of any subtype in a susceptible host animal. For example, the influenza virus can be an HA subtype of H1, H2, H3, H4, H5, H6, H7, H8, H9, H10, H11, H12, H13, H14, H15, or H16, and an NA subtype of N1, N2, N3, N4, N5, N6, N7, N8, or N9. In one embodiment, the HA subtype is H3 or H5. In a further embodiment, the NA subtype is N7 or N8. In a specific embodiment, an immune response is induced against an influenza virus of subtype H3N8. In one embodiment, the host animal is a canid. Canines include wild, zoo, and domestic canines, such as wolves, coyotes, and foxes. Canines also include dogs, particularly domestic dogs, such as, for example, pure-bred and/or mongrel companion dogs, show dogs, working dogs, herding dogs, hunting dogs, guard dogs, police dogs, racing dogs, and/or laboratory dogs. In a specific embodiment, the host animal is a domesticated dog, such as a greyhound. In one embodiment, an animal is administered an effective amount of an immunogenic composition of the present invention sufficient to induce an immune response against an influenza virus of the invention. The immune response can be a humoral and/or cellular immune response. In a specific embodiment, the immune response is a protective immune response that is capable of preventing or minimizing viral infection in the immunized host animal for some period of time subsequent to the immunization. Thus, the subject invention also concerns vaccine compositions and methods that can provide a vaccinated animal with a protective immune response to a virus of the present invention.


As described herein, the vaccine or immunogenic compositions of the subject invention may comprise cell-free whole virus, including attenuated or inactivated virus, or portions of the virus, including subvirion particles (including “split vaccine” wherein a virion is treated to remove some or all viral lipids), viral proteins (including individual proteins and macromolecular complexes of multiple proteins), polypeptides, and peptides, as well as virus-infected cell lines, or a combination of any of these. Vaccine or immunogenic compositions comprising virus-infected cell lines may comprise multiple cell lines, each infected with a different viral strain.


In one embodiment of the invention, a canine may be immunized with one or more inactivated (i.e., killed) and/or live attenuated influenza virus vaccines or vaccines comprising one or a multiplicity of influenza virus antigens from one or more virus isolates. In one embodiment, the influenza virus is a canine influenza virus. In another embodiment, the influenza virus is an equine influenza virus that encodes or expresses a polypeptide that has at least about 90%, or at least about 95%, or at least about 96%, or 97%, or 98%, or 99% or more amino acid sequence identity with a canine influenza virus polypeptide. In one embodiment, an influenza antigen used in a vaccine of the present invention has at least about 96% sequence identity with an HA antigen and/or NA antigen of a canine influenza virus.


An example of an inactivated vaccine is EQUICINE II™, which has been marketed by Intervet Inc. (Millsboro, DE, USA) as a liquid vaccine. EQUICINE II™ contains inactivated A/Pennsylvania/63 influenza virus (“A/Pa/63”) and A/equine/Kentucky/93 influenza virus (“A/KY/93”) with carbopol (i.e., HAVLOGEN® (Intervet Inc.)). More specifically, a dose of EQUICINE II™ contains: inactivated A/Pa/63 at 106.0 EID50, inactivated A/KY/93 at 106.7 EID50, 0.25% by volume carbopol, and sufficient PBS to create a total volume of 1 ml.


Another example of an inactivated vaccine is equine flu virus A/equine/Ohio/03 (“Ohio 03”). In some embodiments, such a vaccine contains CARBIGEN™, which is an emulsified polymer-based adjuvant commercially available from MVP Laboratories, Inc. (Ralston, NE) such vaccines, a dosage unit typically comprises at least about 250 HA units of the virus, from about 250 to about 12,500 HA units of the virus, or from about 1000 to about 6200 HA units of the virus. The recommended concentration of CARBIGEN™ is from about 5 to about 30% (by mass).


An example of a live attenuated vaccine is modified live equine/Kentucky/91 (“A/KY/91”) influenza in the form of a freeze-dried vaccine that may be reconstituted with water. In some embodiments, this reconstitution is conducted using vaccine-grade water sufficient to bring the vaccine dosage to a total volume of 1 ml. Aspects of such vaccines are discussed in, for example, U.S. Pat. Nos. 6,436,408; 6,398,774; and 6,177,082, which are incorporated by reference in their entirety into this patent. When reconstituted, a dose of such a vaccine may, for example, contain A/KY/91 at 107.2 TCID50 per ml, 0.015 grams N-Z AMINE AS™ per ml, 0.0025 grams gelatin per ml, and 0.04 grams D lactose per ml. N-Z AMINE AS™ is a refined source of amino acids and peptides produced by enzymatic hydrolysis of casein. N-Z AMINE AS™ is marketed by Kerry Bio-Science (Norwich, NY, USA).


In a preferred embodiment, the vaccine comprises an H3 influenza antigen having at least about 93% homology with Florida/43/2004 in HA coding sequences, such as, for example, the equine/New Market/79 strain. Preferred homology is at least about 96%, such as, for example, the equine/Alaska/1/91 and equine/Santiago/85 strains. In the examples that follow, the equine/Kentucky/91, equine-2/Kentucky/93, equine-1/Pennsylvania/63, and equine Ohio/03 influenza antigens were incorporated into vaccines. Preferred vaccines also include vaccines comprising equine/Wisconsin/03, equine/Kentucky/02, equine/Kentucky/93, and equine/New Market 2/93. In the examples that follow, H3N8 viruses were used. It is believed, however, that other H3 influenza viruses can be used in accordance with this invention.


Live attenuated vaccines can be prepared by conventional means. Such means generally include, for example, modifying pathogenic strains by in vitro passaging, cold adaptation, modifying the pathogenicity of the organism by genetic manipulation, preparation of chimeras, insertion of antigens into viral vectors, selecting non-virulent wild type strains, etc.


In some embodiments, the live attenuated virus strain is derived by serial passage of the wild-type virus through cell culture, laboratory animals, non-host animals, or eggs. The accumulation of genetic mutation during such passage(s) typically leads to progressive loss of virulence of the organism to the original host.


In some embodiments, the live attenuated virus strain is prepared by co-infection of permissible cells with an attenuated mutant virus and pathogenic virus. The desired resultant recombinant virus has the safety of the attenuated virus with genes coding for protective antigens from the pathogenic virus.


In some embodiments, the live attenuated virus strain is prepared by cold adaptation. A cold-adapted virus has an advantage of replicating only at the temperature found in upper respiratory tract. A method of generation of a cold-adapted equine influenza virus has been described in U.S. Pat. No. 6,177,082. A desired resulting cold-adapted virus confers one or more of the following phenotypes: cold adaptation, temperature sensitivity, dominant interference, and/or attenuation.


In some embodiments, the live attenuated virus strain is prepared by molecular means, such as point mutation, deletion, or insertion to convert a pathogenic virus to a non-pathogenic or less-pathogenic virus compared to the original virus, while preserving the protective properties of the original virus.


In some embodiments, the live attenuated virus is prepared by cloning the candidate of genes of protective antigens into a genome of a non-pathogenic or less-pathogenic virus or other organism.


Inactivated (i.e., “killed”) virus vaccines may be prepared by inactivating the virus using conventional methods. Typically, such vaccines include excipients that may enhance an immune response, as well as other excipients that are conventionally used in vaccines. For example, in the examples that follow, EQUICINE II™ comprises HAVLOGEN®. Inactivation of the virus can be accomplished by treating the virus with inactivation chemicals (e.g., formalin, beta propiolactone (“BPL”), bromoethylamine (“BEA”), and binary ethylenimine (“BEI”)) or by non-chemical methods (e.g., heat, freeze/thaw, or sonication) to disable the replication capacity of the virus.


In the examples that follow, equine/Ohio/03 was used as a challenge virus. It is known to have about 99% homology with Florida/43/04 isolates, and has been shown to induce symptoms of infection and seroconversion in dogs. Example 18 illustrates the efficacy of equine influenza vaccine in dogs, showing hemagglutination inhibition (or “HI” or “HAI”) titers in dogs vaccinated with inactivated Ohio 03 antigen in a vaccine composition comprising CARBIGEN™ adjuvant. Table 29 shows titers pre-vaccination, post-vaccination, and post-second vaccination, as well as post-challenge. The results indicate HI titers at each stage post-vaccination for the vaccinated dogs, with little or no increase for controls. Table 30 illustrates the clinical signs, virus isolation, and histopathology results from the same study. Although challenged animals did not show clinical signs, virus shedding, or positive histopathology, the positive HI titers (Table 29) indicate significant antibody titers in immunized animals.


It should be noted that other H3 influenza virus antigen vaccines are encompassed by this invention as well. Those described in this specification and the following examples are provided to illustrate the invention and its preferred embodiments, and not to limit the scope of the invention claimed.


It should further be noted that influenza antigens other than H3 influenza virus antigens may be used in accordance with this invention. Such antigens include, for example, those from equine/PA/63, which is an equine A1 subtype (H7N7). It is contemplated that one or more of such antigens may be used with or without one or more H3 influenza antigens.


In general, the vaccine is administered in a therapeutically effective amount. A “therapeutically effective amount” is an amount sufficient to induce a protective response in the canine patient against the target virus. Typically, a dosage is “therapeutically effective” if it prevents, reduces the risk of, delays the onset of, reduces the spread of, ameliorates, suppresses, or eradicates the influenza or one or more (typically two or more) of its symptoms. Typical influenza symptoms include, for example, fever (for dogs, typically ≥103.0° F.; ≥39.4° C.), cough, sneezing, histopathological lesions, ocular discharge, nasal discharge, vomiting, diarrhea, depression, weight loss, gagging, hemoptysis, and/or audible rales. Other often more severe symptoms may include, for example, hemorrhage in the lungs, mediastanum, or pleural cavity; tracheitis; bronchitis; bronchiolitis; supportive bronchopneumonia; and/or infiltration of the epithelial lining and airway lumens of the lungs with neutrophils and/or macrophages.


The vaccine may be administered as part of a combination therapy, i.e., a therapy that includes, in addition to the vaccine itself, administering one or more additional active agents, adjuvants, therapies, etc. In that instance, it should be recognized the amount of vaccine that constitutes a “therapeutically effective” amount may be less than the amount of vaccine that would constitute a “therapeutically effective” amount if the vaccine were to be administered alone. Other therapies may include those known in the art, such as, for example, anti-viral medications, analgesics, fever-reducing medications, expectorants, anti-inflammation medications, antihistamines, antibiotics to treat bacterial infection that results from the influenza virus infection, rest, and/or administration of fluids. In some embodiments, the vaccine of this invention is administered in combination with a bordetella vaccine, adenovirus vaccine, and/or parainfluenza virus vaccine.


In some embodiments, for example, a typical dose for a live attenuated vaccine is at least about 103 pfu/canine, and more typically from about 103 to about 109 pfu/canine. In this patent, “pfu” means “plaque forming units”. In some embodiments, a typical dose for a live attenuated vaccine is at least about 103 TCID50/canine, and more typically from about 103 to about 109 TCID50/canine. In some embodiments, a typical dose for a live attenuated vaccine is at least about 103 EID50/canine, and more typically from about 103 to about 109 EID50/canine. In some embodiments, a typical dose for a killed vaccine is at least about 40 HA units, typically from about 40 to about 10,000 HA units, and more typically from about 500 to about 6200 HA units. In some embodiments, the dose is from about 6100 to about 6200 HA units.


In some preferred embodiments, the vaccine comprises a live attenuated vaccine at a concentration which is at least about 100.5 pfu/canine greater than the immunogenicity level. In some preferred embodiments, the vaccine comprises a live attenuated vaccine at a concentration which is at least about 100.5 TCID50/canine greater than the immunogenicity level. In some preferred embodiments, the vaccine comprises a live attenuated vaccine at a concentration which is at least about 1005 EID50/canine greater than the immunogenicity level.


The immunogenicity level may be determined experimentally by challenge dose titration study techniques generally known in the art. Such techniques typically include vaccinating a number of canines with the vaccine at different dosages, and then challenging the canines with the virulent virus to determine the minimum protective dose.


Factors affecting the preferred dosage regimen may include, for example, the type (e.g., species and breed), age, weight, sex, diet, activity, lung size, and condition of the subject; the route of administration; the efficacy, safety, and duration-of-immunity profiles of the particular vaccine used; whether a delivery system is used; and whether the vaccine is administered as part of a drug and/or vaccine combination. Thus, the dosage actually employed can vary for specific animals, and, therefore, can deviate from the typical dosages set forth above. Determining such dosage adjustments is generally within the skill of those in the art using conventional means. It should further be noted that live attenuated viruses are generally self-propagating; thus, the specific amount of such a virus administered is not necessarily critical.


It is contemplated that the vaccine may be administered to the canine patient a single time; or, alternatively, two or more times over days, weeks, months, or years. In some embodiments, the vaccine is administered at least two times. In some such embodiments, for example, the vaccine is administered twice, with the second dose (e.g., the booster) being administered at least about 2 weeks after the first. In some embodiments, the vaccine is administered twice, with the second dose being administered no greater than 8 weeks after the first. In some embodiments, the second dose is administered at from about 2 weeks to about 4 years after the first dose, from about 2 to about 8 weeks after the first dose, or from about 3 to about 4 weeks after the first dose. In some embodiments, the second dose is administered about 4 weeks after the first dose. In the above embodiments, the first and subsequent dosages may vary, such as, for example, in amount and/or form. Often, however, the dosages are the same as to amount and form. When only a single dose is administered, the amount of vaccine in that dose alone generally comprises a therapeutically effective amount of the vaccine. When, however, more than one dose is administered, the amounts of vaccine in those doses together may constitute a therapeutically effective amount.


In some embodiments, the vaccine is administered before the canine recipient is infected with influenza. In such embodiments, the vaccine may, for example, be administered to prevent, reduce the risk of, or delay the onset of influenza or one or more (typically two or more) influenza symptoms.


In some embodiments, the vaccine is administered after the canine recipient is infected with influenza. In such embodiments, the vaccine may, for example, ameliorate, suppress, or eradicate the influenza or one or more (typically two or more) influenza symptoms.


The preferred composition of the vaccine depends on, for example, whether the vaccine is an inactivated vaccine, live attenuated vaccine, or both. It also depends on the method of administration of the vaccine. It is contemplated that the vaccine will comprise one or more conventional pharmaceutically acceptable carriers, adjuvants, other immune-response enhancers, and/or vehicles (collectively referred to as “excipients”). Such excipients are generally selected to be compatible with the active ingredient(s) in the vaccine. Use of excipients is generally known to those skilled in the art.


The term “pharmaceutically acceptable” is used adjectivally to mean that the modified noun is appropriate for use in a pharmaceutical product. When it is used, for example, to describe an excipient in a pharmaceutical vaccine, it characterizes the excipient as being compatible with the other ingredients of the composition and not disadvantageously deleterious to the intended recipient canine.


The vaccines may be administered by conventional means, including, for example, mucosal administration, (such as intranasal, oral, intratracheal, and ocular), and parenteral administration. Mucosal administration is often particularly advantageous for live attenuated vaccines. Parenteral administration is often particularly advantageous for inactivated vaccines.


Mucosal vaccines may be, for example, liquid dosage forms, such as pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Excipients suitable for such vaccines include, for example, inert diluents commonly used in the art, such as, water, saline, dextrose, glycerol, lactose, sucrose, starch powder, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinylpyrrolidone, and/or polyvinyl alcohol. Excipients also can comprise various wetting, emulsifying, suspending, flavoring (e.g., sweetening), and/or perfuming agents.


Oral mucosal vaccines also may, for example, be tableted or encapsulated for convenient administration. Such capsules or tablets can contain a controlled-release formulation. In the case of capsules, tablets, and pills, the dosage forms also can comprise buffering agents, such as sodium citrate, or magnesium or calcium carbonate or bicarbonate. Tablets and pills additionally can be prepared with enteric coatings.


It is contemplated that the vaccine may be administered via the canine patient's drinking water and/or food. It is further contemplated that the vaccine may be administered in the form of a treat or toy.


“Parenteral administration” includes subcutaneous injections, submucosal injections, intravenous injections, intramuscular injections, intrasternal injections, transcutaneous injections, and infusion. Injectable preparations (e.g., sterile injectable aqueous or oleaginous suspensions) can be formulated according to the known art using suitable excipients, such as vehicles, solvents, dispersing, wetting agents, emulsifying agents, and/or suspending agents. These typically include, for example, water, saline, dextrose, glycerol, ethanol, corn oil, cottonseed oil, peanut oil, sesame oil, benzyl alcohol, benzyl alcohol, 1,3-butanediol, Ringer's solution, isotonic sodium chloride solution, bland fixed oils (e.g., synthetic mono- or diglycerides), fatty acids (e.g., oleic acid), dimethyl acetamide, surfactants (e.g., ionic and non-ionic detergents), propylene glycol, and/or polyethylene glycols. Excipients also may include small amounts of other auxiliary substances, such as pH buffering agents.


The vaccine may include one or more excipients that enhance a canine patient's immune response (which may include an antibody response, cellular response, or both), thereby increasing the effectiveness of the vaccine. Use of such excipients (or “adjuvants”) may be particularly beneficial when using an inactivated vaccine. The adjuvant(s) may be a substance that has a direct (e.g., cytokine or Bacille Calmette-Guerin (“BCG”)) or indirect effect (liposomes) on cells of the canine patient's immune system. Examples of often suitable adjuvants include oils (e.g., mineral oils), metallic salts (e.g., aluminum hydroxide or aluminum phosphate), bacterial components (e.g., bacterial liposaccharides, Freund's adjuvants, and/or MDP), plant components (e.g., Quil A), and/or one or more substances that have a carrier effect (e.g., bentonite, latex particles, liposomes, and/or Quil A, ISCOM). As noted above, adjuvants also include, for example, CARBIGEN™ and carbopol. It should be recognized that this invention encompasses both vaccines that comprise an adjuvant(s), as well as vaccines that do not comprise any adjuvant.


It is contemplated that the vaccine may be freeze-dried (or otherwise reduced in liquid volume) for storage, and then reconstituted in a liquid before or at the time of administration. Such reconstitution may be achieved using, for example, vaccine-grade water.


The present invention further comprises kits that are suitable for use in performing the methods described above. The kit comprises a dosage form comprising a vaccine described above. The kit also comprises at least one additional component, and, typically, instructions for using the vaccine with the additional component(s). The additional component(s) may, for example, be one or more additional ingredients (such as, for example, one or more of the excipients discussed above, food, and/or a treat) that can be mixed with the vaccine before or during administration. The additional component(s) may alternatively (or additionally) comprise one or more apparatuses for administering the vaccine to the canine patient. Such an apparatus may be, for example, a syringe, inhaler, nebulizer, pipette, forceps, or any medically acceptable delivery vehicle. In some embodiments, the apparatus is suitable for subcutaneous administration of the vaccine. In some embodiments, the apparatus is suitable for intranasal administration of the vaccine.


Other excipients and modes of administration known in the pharmaceutical or biologics arts also may be used.


The vaccine or immunogenic compositions of the subject invention also encompass recombinant viral vector-based constructs that may comprise, for example, genes encoding HA protein, NA protein, nucleoprotein, polymerase basic protein, polymerase acidic protein, and/or matrix protein of an influenza virus of the present invention. Any suitable viral vector that can be used to prepare a recombinant vector/virus construct is contemplated for use with the subject invention. For example, viral vectors derived from adenovirus, avipox, herpesvirus, vaccinia, canarypox, entomopox, swinepox, West Nile virus and others known in the art can be used with the compositions and methods of the present invention. Recombinant polynucleotide vectors that encode and express components can be constructed using standard genetic engineering techniques known in the art. In addition, the various vaccine compositions described herein can be used separately and in combination with each other. For example, primary immunizations of an animal may use recombinant vector-based constructs, having single or multiple strain components, followed by secondary boosts with vaccine compositions comprising inactivated virus or inactivated virus-infected cell lines. Other immunization protocols with the vaccine compositions of the invention are apparent to persons skilled in the art and are contemplated within the scope of the present invention.


The subject invention also concerns reassortant virus comprising at least one gene or genomic segment of an influenza virus of the present invention and the remainder of viral genes or genomic segments from a different influenza virus of the invention or from an influenza virus other than a virus of the present invention. Reassortant virus can be produced by genetic reassortant of nucleic acid of a donor influenza virus of the present invention with nucleic acid of a recipient influenza virus and then selecting for reassortant virus that comprises the nucleic acid of the donor virus. Methods to produce and isolate reassortant virus are well known in the art (Fields et al., 1996). In one embodiment, a reassortant virus of the invention comprises genes or genomic segments of a human, avian, swine, or equine influenza virus. A reassortant virus of the present invention can include any combination of nucleic acid from donor and recipient influenza virus so long as the reassortant virus comprises at least one gene or genomic segment from a donor influenza virus of the present invention. In one embodiment, a recipient influenza virus can be an equine influenza virus.


Natural, recombinant or synthetic polypeptides of viral proteins, and peptide fragments thereof, can also be used as vaccine compositions according to the subject methods. In one embodiment, a vaccine composition comprises a polynucleotide or a polypeptide of a canine influenza virus. In one embodiment, a vaccine composition comprises a polynucleotide encoding a polypeptide having the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a specific embodiment, the polynucleotide encoding the amino acid sequence shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, comprises the nucleotide sequence shown in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, respectively, or a sequence encoding a functional and/or immunogenic fragment or variant of any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78. In a further specific embodiment, a polynucleotide of the invention can comprise: Nucleotides 1-2271 of SEQ ID NO: 3; Nucleotides 1-2148 of SEQ ID NO: 5; Nucleotides 1-657 of SEQ ID NO: 7; Nucleotides 1-1494 of SEQ ID NO: 9; Nucleotides 1-1410 of SEQ ID NO: 11; Nucleotides 1-756 of SEQ ID NO: 13; Nucleotides 1-1695 of SEQ ID NO: 15; Nucleotides 1-2271 of SEQ ID NO: 19; Nucleotides 1-2148 of SEQ ID NO: 21; Nucleotides 1-657 of SEQ ID NO: 23; Nucleotides 1-1494 of SEQ ID NO: 25; Nucleotides 1-756 of SEQ ID NO: 29; Nucleotides 1-1695 of SEQ ID NO: 31; Nucleotides 1-2277 of SEQ ID NO: 47; Nucleotides 1-2271 of SEQ ID NO: 49; Nucleotides 1-2148 of SEQ ID NO: 51; Nucleotides 1-690 of SEQ ID NO: 53; Nucleotides 1-1494 of SEQ ID NO: 55; Nucleotides 1-1410 of SEQ ID NO: 57; Nucleotides 1-756 of SEQ ID NO: 59; Nucleotides 1-1695 of SEQ ID NO: 61; Nucleotides 1-2277 of SEQ ID NO: 63; Nucleotides 1-2271 of SEQ ID NO: 65; Nucleotides 1-2148 of SEQ ID NO: 67; Nucleotides 1-690 of SEQ ID NO: 69; Nucleotides 1-1494 of SEQ ID NO: 71; Nucleotides 1-1410 of SEQ ID NO: 73; Nucleotides 1-756 of SEQ ID NO: 75; and Nucleotides 1-1695 of SEQ ID NO: 77. In another embodiment, a vaccine composition comprises a polypeptide having the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a further embodiment, a vaccine composition comprises a polynucleotide or a polypeptide of an equine influenza virus wherein the polynucleotide or polypeptide has at least about 90%, or at least about 95%, or at least about 96%, or 97%, or 98%, or 99% or more sequence identity with a canine influenza polynucleotide or polypeptide. In one embodiment, viral polypeptides derived from multiple strains can be combined in a vaccine composition and are used to vaccinate a host animal. For example, polypeptides based on the viral HA protein from at least two different strains of influenza virus of the invention can be combined in the vaccine. The polypeptides may be homologous to one strain or may comprise Ahybrid@ or Achimeric@ polypeptides whose amino acid sequence is derived from joining or linking polypeptides from at least two distinct strains. Procedures for preparing viral polypeptides are well known in the art. For example, viral polypeptides and peptides can be synthesized using solid-phase synthesis methods (Merrifield, 1963). Viral polypeptides and peptides can also be produced using recombinant DNA techniques wherein a polynucleotide molecule encoding an viral protein or peptide is expressed in a host cell, such as bacteria, yeast, or mammalian cell lines, and the expressed protein purified using standard techniques of the art.


Vaccine compositions of the present invention also include naked nucleic acid compositions. In one embodiment, a nucleic acid may comprise a nucleotide sequence encoding an HA and/or an NA protein of an influenza virus of the present invention. Methods for nucleic acid vaccination are known in the art and are described, for example, in U.S. Pat. Nos. 6,063,385 and 6,472,375. The nucleic acid can be in the form of a plasmid or a gene expression cassette. In one embodiment, the nucleic acid is provided encapsulated in a liposome which is administered to an animal.


Vaccine compositions and immunogens, such as polypeptides and nucleic acids, that can be used in accordance with the present invention can be provided with a pharmaceutically-acceptable carrier or diluent. Compounds and compositions useful in the subject invention can be formulated according to known methods for preparing pharmaceutically useful compositions. Formulations are described in detail in a number of sources which are well known and readily available to those skilled in the art. For example, Remington's Pharmaceutical Science by E. W. Martin, Easton Pennsylvania, Mack Publishing Company, 19th ed., 1995, describes formulations which can be used in connection with the subject invention. In general, the compositions of the subject invention will be formulated such that an effective amount of an immunogen is combined with a suitable carrier in order to facilitate effective administration of the composition. The compositions used in the present methods can also be in a variety of forms. These include, for example, solid, semi-solid, and liquid dosage forms, such as tablets, pills, powders, liquid solutions or suspension, suppositories, injectable and infusible solutions, and sprays. The preferred form depends on the intended mode of administration and therapeutic application. The compositions also preferably include conventional pharmaceutically acceptable carriers and diluents which are known to those skilled in the art. Examples of carriers or diluents for use with the subject peptidomimetics include, but are not limited to, water, saline, oils including mineral oil, ethanol, dimethyl sulfoxide, gelatin, cyclodextrans, magnesium stearate, dextrose, cellulose, sugars, calcium carbonate, glycerol, alumina, starch, and equivalent carriers and diluents, or mixtures of any of these. Formulations of an immunogen of the invention can also comprise suspension agents, protectants, lubricants, buffers, preservatives, and stabilizers. To provide for the administration of such dosages for the desired therapeutic treatment, pharmaceutical compositions of the invention will advantageously comprise between about 0.1% and 45%, and especially, 1 and 15% by weight of the immunogen or immunogens based on the weight of the total composition including carrier or diluent.


The vaccine and immunogenic compositions of the subject invention can be prepared by procedures well known in the art. For example, the vaccine or immunogens are typically prepared as injectables, e.g., liquid solutions or suspensions. The vaccine or immunogens are administered in a manner that is compatible with dosage formulation, and in such amount as will be therapeutically effective and immunogenic in the recipient. The optimal dosages and administration patterns for a particular vaccine or immunogens formulation can be readily determined by a person skilled in the art.


Peptides and/or polypeptides of the present invention can also be provided in the form of a multiple antigenic peptide (MAP) construct. The preparation of MAP constructs has been described in Tam (1988). MAP constructs utilize a core matrix of lysine residues onto which multiple copies of an immunogen are synthesized (Posnett et al., 1988). Multiple MAP constructs, each containing the same or different immunogens, can be prepared and administered in a vaccine composition in accordance with methods of the present invention. In one embodiment, a MAP construct is provided with and/or administered with one or more adjuvants. Influenza polypeptides of the invention can also be produced and administered as macromolecular protein structures comprising one or more polypeptides. Published U.S. Patent Application US2005/0009008 describes methods for producing virus-like particles as a vaccine for influenza virus.


According to the methods of the subject invention, the vaccine and immunogenic compositions described herein are administered to susceptible hosts, typically canids, and more typically domesticated dogs, in an effective amount and manner to induce protective immunity against subsequent challenge or infection of the host by virus. In one embodiment, the host animal is a canid. Canines include wild, zoo, and domestic canines, such as wolves, coyotes, and foxes. Canines also include dogs, particularly domestic dogs, such as, for example, pure-bred and/or mongrel companion dogs, show dogs, working dogs, herding dogs, hunting dogs, guard dogs, police dogs, racing dogs, and/or laboratory dogs. In a specific embodiment, the host animal is a domesticated dog, such as a greyhound. The vaccines or immunogens are typically administered parenterally, by injection, for example, either subcutaneously, intraperitoneally, or intramuscularly. Other suitable modes of administration include oral or nasal administration. Usually, the vaccines or immunogens are administered to an animal at least two times, with an interval of one or more weeks between each administration. However, other regimens for the initial and booster administrations of the vaccine or immunogens are contemplated, and may depend on the judgment of the practitioner and the particular host animal being treated.


Virus and virus-infected cells in a vaccine formulation may be inactivated or attenuated using methods known in the art. For example, whole virus and infected cells can be inactivated or attenuated by exposure to paraformaldehyde, formalin, beta propiolactone (BPL), bromoethylamine (BEA), binary ethylenimine (BEI), phenol, UV light, elevated temperature, freeze thawing, sonication (including ultrasonication), and the like. The amount of cell-free whole virus in a vaccine dose can be in the range from about 0.1 mg to about 5 mg, and more usually being from about 0.2 mg to about 2 mg. The dosage for vaccine formulations comprising virus-infected cell lines will usually contain from about 106 to about 108 cells per dose, and more usually from about 5×106 to about 7.5×107 cells per dose. The amount of protein or peptide immunogen in a dose for an animal can vary from about 0.1 μg to 10000 μg, or about 1 μg to 5000 μg, or about 10 μg to 1000 μg or about 25 μg to 750 μg, or about 50 μg to 500 μg or 100 μg to 250 μg, depending upon the size, age, etc., of the animal receiving the dose.


An immunogenic or vaccine composition of the invention, such as virus or virus-infected cells or viral proteins or peptides, can be combined with an adjuvant, typically just prior to administration. Adjuvants contemplated for use in the vaccine formulations include threonyl muramyl dipeptide (MDP) (Byars et al., 1987), saponin, Cornebacterium parvum, Freund's complete and Fruend's incomplete adjuvants, aluminum, or a mixture of any of these. A variety of other adjuvants suitable for use with the methods and vaccines of the subject invention, such as alum, are well known in the art and are contemplated for use with the subject invention.


The subject invention also concerns antibodies that bind specifically to a protein or a peptide of the present invention. Antibodies of the subject invention include monoclonal and polyclonal antibody compositions. Preferably, the antibodies of the subject invention are monoclonal antibodies. Whole antibodies and antigen binding fragments thereof are contemplated in the present invention. Thus, for example, suitable antigen binding fragments include Fab2, Fab and Fv antibody fragments. Antibodies of the invention can be labeled with a detectable moiety, such as a fluorescent molecule (e.g., fluorescein or an enzyme). The subject invention also concerns methods and compositions for detection and identification of an influenza virus of the invention and for diagnosis of infection of an animal with an influenza virus of the present invention. The methods of the invention include detection of the presence of canine influenza, in a biological sample from an animal. The detection of canine influenza in a sample, is useful to diagnose canine influenza in an animal. In turn, this information can provide the ability to determine the prognosis of an animal based on distinguishing levels of canine influenza present over time, and can assist in selection of therapeutic agents and treatments for the animal, and assist in monitoring therapy. The method also provides the ability to establish the absence of canine influenza in an animal tested.


The ability to detect canine influenza in an animal permits assessment of outbreaks of canine influenza in different geographical locations. This information also permits early detection so that infected animals can be isolated, to limit the spread of disease, and allows early intervention for treatment options. In addition, having this information available can provide direction to medical personnel for preparing to treat large numbers of ill animals, including assembling medical supplies, and, if available, vaccines.


In one embodiment, a method of the present invention involves the collection of a biological sample from a test animal, such as a canine. The biological sample may be any biological material, including, cells, tissue, hair, whole blood, serum, plasma, nipple aspirate, lung lavage, cerebrospinal fluid, saliva, sweat and tears.


The animal test sample may come from an animal suspected of having canine influenza virus, whether or not the animal exhibits symptoms of the disease. Control samples can also be provided or collected from animals known to be free of canine influenza. Additional controls may be provided, e.g., to reduce false positive and false negative results, and verify that the reagents in the assay are actively detecting canine influenza A virus.


In addition to detecting the presence or absence of canine influenza in a biological sample, the methods of detection used in the invention can detect mutations in canine influenza virus, such as changes in nucleic acid sequence, that may result from the environment, drug treatment, genetic manipulations or mutations, injury, change in diet, aging, or any other characteristic(s) of an animal. Mutations may also cause canine influenza A to become resistant to a drug that was formerly effective, or to enable the virus to infect and propagate in a different species of animal, or human. For example, avian influenza A virus has been shown to infect other animals and humans.


In one embodiment for detecting an influenza virus in an animal, diagnosis is facilitated by the collection of high-quality specimens, their rapid transport to a testing facility, and appropriate storage, before laboratory testing. Virus is best detected in specimens containing infected cells and secretions. In one embodiment, specimens for the direct detection of viral antigens and/or for nucleic acids and/or virus isolation in cell cultures are taken during the first 3 days after onset of clinical symptoms. A number of types of specimens are suitable to diagnose virus infections of the upper respiratory tract, including, but not limited to, nasal swab, nasopharyngeal swab, nasopharyngeal aspirate, nasal wash and throat swabs. In addition to swabs, samples of tissue or serum may be taken, and invasive procedures can also be performed.


In one embodiment, respiratory specimens are collected and transported in 1-5 ml of virus transport media. A number of media that are satisfactory for the recovery of a wide variety of viruses are commercially available. Clinical specimens are added to transport medium. Nasal or nasopharyngeal swabs can also be transported in the virus transport medium. One example of a transport medium is 10 gm of veal infusion broth and 2 gm of bovine albumin fraction V, added to sterile distilled water to 400 m. Antibiotics such as 0.8 ml gentamicin sulfate solution (50 mg/ml) and 3.2 ml amphotericin B (250 μg/ml) can also be added. The medium is preferably sterilized by filtration. Nasal washes, such as sterile saline (0.85% NaCl), can also be used to collect specimens of respiratory viruses.


In one embodiment, sera is collected in an amount of from 1-5 ml of whole blood from an acute-phase animal, soon after the onset of clinical symptoms, and preferably not later than 7 days. A convalescent-phase serum specimen can also be collected, for example at about 14 days after onset of symptoms. Serum specimens can be useful for detecting antibodies against respiratory viruses in a neutralization test.


In some instances, samples may be collected from individual animals over a period of time (e.g., once a day, once a week, once a month, biannually or annually). Obtaining numerous samples from an individual animal, over a period of time, can be used to verify results from earlier detections, and/or to identify response or resistance to a specific treatment, e.g., a selected therapeutic drug.


The methods of the present invention can be used to detect the presence of one or more pathological agents in a test sample from an animal, and the level of each pathological agent. Any method for detecting the pathological agent can be used, including, but not limited to, antibody assays including enzyme-linked immunosorbent assays (ELISAs), indirect fluorescent antibody (IFA) tests, hemagglutinating, and inhibition of hemagglutination (HI) assays, and Western Blot. Known cell-culture methods can also be used. Positive cultures can be further identified using immunofluorescence of cell cultures or HI assay of the cell culture medium (supernatant).


In addition, methods for detecting nucleic acid (DNA or RNA) or protein can be used. Such methods include, but are not limited to, polymerase chain reaction (PCR), and reverse transcriptase (RT) PCR tests and real time tests, and quantitative nuclease protection assays. There are commercially available test kits available to perform these assays. For example, QIAGEN (Valencia, CA) sells a one step RT-PCR kit, and viral RNA extraction kit.


In one embodiment, the method utilizes an antibody specific for a virus or viral protein of the invention. In a specific embodiment, an antibody specific for an HA protein of a virus of the invention is utilized. In another embodiment, an antibody specific for an NP protein of a virus of the invention is used. A suitable sample, such as from the nasal or nasopharyngeal region, is obtained from an animal and virus or viral protein is isolated therefrom. The viral components are then screened for binding of an antibody specific to a protein, such as HA or NP, of a virus of the invention. In another embodiment, a serum sample (or other antibody containing sample) is obtained from an animal and the serum screened for the presence of antibody that binds to a protein of a virus of the invention. For example, an ELISA assay can be performed where the plate walls have HA and/or NP protein, or a peptide fragment thereof, bound to the wall. The plate wall is then contacted with serum or antibody from a test animal. The presence of antibody in the animal that binds specifically to the HA and/or NP protein is indicative that the test animal is infected or has been infected with an influenza virus of the present invention.


In one embodiment, the presence of a pathological agent is detected by determining the presence or absence of antibodies against the agent, in a biological sample. It can take some time (e.g. months) after an animal is infected before antibodies can be detected in a blood test. Once formed, antibodies usually persist for many years, even after successful treatment of the disease. Finding antibodies to canine influenza A may not indicate whether the infection was recent, or sometime in the past.


Antibody testing can also be done on fluid(s). Antibody assays include enzyme-linked immunosorbent assays (ELISAs), indirect fluorescent antibody (IFA) assays, and Western Blot. Preferably, antibody testing is done using multiple assays, for example ELISA or IFA followed by Western blot. Antibody assays can be done in a two-step process, using either an ELISA or IFA assay, followed by a Western blot assay. ELISA is considered a more reliable and accurate assay than IFA, but IFA may be used if ELISA is not available. The Western blot test (which is a more specific test) can also be done in all animals, particularly those that have tested positive or borderline positive (equivocal) in an ELISA or IFA assay.


Other antibody-based tests that can be used for detection of influenza virus include hemagglutination inhibition assays. Hemagglutination activity can be detected in a biological sample from an animal, using chicken or turkey red blood cells as described (Burleson et al., 1992) and Kendal et al., 1982). In one embodiment, an influenza or an HA protein or peptide of the invention is contacted with a test sample containing serum or antibody. Red blood cells (RBC) from an animal, such as a bird, are then added. If antibody to HA is present, then the RBC will not agglutinate. If antibody to HA is not present, the RBC will agglutinate in the presence of HA. Variations and modifications to standard hemagglutination inhibition assays are known in the art and contemplated within the scope of the present invention.


Infection of an animal can also be determined by isolation of the virus from a sample, such as a nasal or nasopharyngeal swab. Viral isolation can be performed using standard methods, including cell culture and egg inoculation.


In a further embodiment, a nucleic acid-based assay can be used for detection of a virus of the present invention. In one embodiment, a nucleic acid sample is obtained from an animal and the nucleic acid subjected to PCR using primers that will generate an amplification product if the nucleic acid contains a sequence specific to an influenza virus of the present invention. In a specific embodiment, RT-PCR is used in an assay for the subject virus. In an exemplified embodiment, real-time RT-PCR is used to assay for an influenza virus of the invention. PCR, RT-PCR and real-time PCR methods are known in the art and have been described in U.S. Pat. Nos. 4,683,202; 4,683,195; 4,800,159; 4,965,188; 5,994,056; 6,814,934; and in Saiki et al. (1985); Sambrook et al. (1989); Lee et al. (1993); and Livak et al. (1995). In one embodiment, the PCR assay uses oligonucleotides specific for an influenza matrix (MA) gene and/or HA gene. The amplification product can also be sequenced to determine if the product has a sequence of an influenza virus of the present invention. Other nucleic acid-based assays can be used for detection and diagnosis of viral infection by a virus of the invention and such assays are contemplated within the scope of the present invention. In one embodiment, a sample containing a nucleic acid is subjected to a PCR-based amplification using forward and reverse primers where the primers are specific for a viral polynucleotide or gene sequence. If the nucleic acid in the sample is RNA, then RT-PCR can be performed. For real-time PCR, a detectable probe is utilized with the primers.


Primer sets specific for the hemagglutinin (HA) gene of many of the circulating influenza viruses are known, and are continually being developed. The influenza virus genome is single-stranded RNA, and a DNA copy (cDNA) must be made using a reverse transcriptase (RT) polymerase. The amplification of the RNA genome, for example using RT-PCR, requires a pair of oligonucleotide primers, typically designed on the basis of the known HA sequence of influenza A subtypes and of neuraminadase (NM)-1. The primers can be selected such that they will specifically amplify RNA of only one virus subtype. DNAs generated by using subtype-specific primers can be further analyzed by molecular genetic techniques such as sequencing. The test is preferably run with a positive control, or products are confirmed by sequencing and comparison with known sequences. The absence of the target PCR products (i.e, a “negative” result) may not rule out the presence of the virus. Results can then be made available within a few hours from either clinical swabs or infected cell cultures. PCR and RT-PCR tests for influenza A virus are described by Fouchier et al., 2000 and Maertzdorf et al., 2004.


The subject invention also concerns methods for screening for compounds or drugs that have antiviral activity against a virus of the present invention. In one embodiment, cells infected with a virus of the invention are contacted with a test compound or drug. The amount of virus or viral activity following contact is then determined. Those compounds or drugs that exhibit antiviral activity can be selected for further evaluation.


The subject invention also concerns isolated cells infected with an influenza virus of the present invention. In one embodiment, the cell is a canine cell, such as canine kidney epithelial cells.


The subject invention also concerns cells transformed with a polynucleotide of the present invention encoding a polypeptide of the invention. Preferably, the polynucleotide sequence is provided in an expression construct of the invention. More preferably, the expression construct provides for overexpression in the cell of an operably linked polynucleotide of the invention. In one embodiment, the cell is transformed with a polynucleotide sequence comprising a sequence encoding the amino acid sequence shown in any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional fragment or variant thereof. In a specific embodiment, the cell is transformed with a polynucleotide encoding the amino acid sequence shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78 comprises the nucleotide sequence shown in SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, respectively, or a sequence encoding a functional fragment or variant of any of SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78. Thus, the subject invention concerns cells transformed with a polynucleotide sequence comprising the nucleotide sequence shown in any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, or a fragment or variant, including a degenerate variant, of any of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77.


The transformed cell can be a eukaryotic cell, for example, a plant cell, including protoplasts, or the transformed cell can be a prokaryotic cell, for example, a bacterial cell such as E. coli or B. subtilis. Animal cells include human cells, mammalian cells, partially canine cells, avian cells, and insect cells. Plant cells include, but are not limited to, dicotyledonous, monocotyledonous, and conifer cells.


The subject invention also concerns plants, including transgenic plants that express and produce a viral protein or polypeptide of the present invention. Plants, plant tissues, and plant cells transformed with or bred to contain a polynucleotide of the invention are contemplated by the present invention. Preferably, the polynucleotide of the invention is overexpressed in the plant, plant tissue, or plant cell. Plants can be used to produce influenza vaccine compositions of the present invention and the vaccines can be administered through consumption of the plant (see, for example, U.S. Pat. Nos. 5,484,719 and 6,136,320).


The subject invention also concerns kits for detecting a virus or diagnosing an infection by a virus of the present invention. In one embodiment, a kit comprises an antibody of the invention that specifically binds to an influenza virus of the present invention, or an antigenic portion thereof. In another embodiment, a kit comprises one or more polypeptides or peptides of the present invention. In a specific embodiment, the polypeptides have an amino acid sequence shown in any of SEQ ID NOs. 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 33, 34, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, or 78, or a functional and/or immunogenic fragment or variant thereof. In a further embodiment, a kit comprises one or more polynucleotides or oligonucleotides of the present invention. In a specific embodiment, the polynucleotides have a nucleotide sequence shown in any of SEQ ID NOs. 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, or 77, or a fragment or variant thereof. A kit may optionally comprise one or more control antibody, control polypeptide or peptide, and/or control polynucleotide or oligonucleotide. The antibody, polypeptides, peptides, polynucleotides, and/or oligonucleotides of the kit can be provided in a suitable container or package.


The subject application also concerns the use of mongrel dogs as a model for infection and pathogenesis of influenza virus. In one embodiment, a mongrel dog is inoculated with an influenza virus, such as a canine influenza virus of the present invention. Optionally, the dog can be administered therapeutic agents subsequent to inoculation. The dog can also have been administered a composition for generating an immune response against an influenza virus prior to inoculation with virus. Tissue, blood, serum, and other biological samples can be obtained before and/or after inoculation and examined for the presence of virus and pathogenesis of tissue using methods known in the art including, but not limited to, PCR, RT-PCR, nucleic acid sequencing, and immunohistochemistry.


Canine influenza virus strains (designated as “A/canine/Florida/43/2004” (ATCC Accession No. PTA-7914) and “A/canine/Florida/242/2003” (ATCC Accession No. PTA-7915)) were deposited with American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, VA 20108, on Oct. 9, 2006. Canine influenza virus strains (designated as “canine/Jax/05” (ATCC Accession No. PTA 7941) and “canine/Miami/05” (ATCC Accession No. PTA-7940)), were deposited with American Type Culture Collection (ATCC), P.O. Box 1549, Manassas, VA 20108, on Oct. 17, 2006. The subject virus strains have been deposited under conditions that assure that access to the cultures will be available during the pendency of this patent application to one determined by the Commissioner of Patents and Trademarks to be entitled thereto under 37 CFR 1.14 and 35 U.S.C. 122. The deposit will be available as required by foreign patent laws in countries wherein counterparts of the subject application, or its progeny, are filed. However, it should be understood that the availability of a deposit does not constitute a license to practice the subject invention in derogation of patent rights granted by governmental action.


Further, the subject virus deposits will be stored and made available to the public in accord with the provisions of the Budapest Treaty for the Deposit of Microorganisms, i.e., it will be stored with all the care necessary to keep it viable and uncontaminated for a period of at least five years after the most recent request for the furnishing of a sample of the deposit, and in any case, for a period of at least thirty (30) years after the date of deposit or for the enforceable life of any patent which may issue disclosing the culture. The depositor acknowledges the duty to replace the deposit should the depository be unable to furnish a sample when requested, due to the condition of the deposit. All restrictions on the availability to the public of the subject culture deposit will be irrevocably removed upon the granting of a patent disclosing it.


Table 57 illustrates the similarities among the amino acid sequences encoded by the hemagglutinin (or “HA”), neuraminidase (or “NA”), and nucleoprotein (NP) genes of the canine influenza virus identified as A/canine/Florida/43/2004 (Ca/Fla/43/04) with H3N8 equine isolates, as well as the canine/Florida/242/2003 isolate.


Any element of any embodiment disclosed herein can be combined with any other element or embodiment disclosed herein and such combinations are specifically contemplated within the scope of the present invention.


All patents, patent applications, provisional applications, and publications referred to or cited herein are incorporated by reference in their entirety, including all figures and tables, to the extent they are not inconsistent with the explicit teachings of this specification.


Materials and Methods for Examples 1-6

Blood and Nasal Swab Collection from Greyhounds.


Acute and convalescent blood samples were collected by jugular venipuncture from clinically diseased or normal greyhounds in racing kennels experiencing outbreaks of respiratory disease. Convalescent samples were collected 4 to 12 weeks after the acute sample. Serum was harvested and stored at −80° C. Nasal swabs were collected and placed in Amies transport medium with charcoal (Becton Dickinson Biosciences) pending submission for bacterial isolation.


Postmortem Examination of Greyhounds.


Complete postmortem examinations were performed by the Anatomic Pathology Service at the University of Florida College of Veterinary Medicine (UF CVM) on 5 of the 8 greyhounds that died in the January 2004 outbreak at a Florida track. Postmortem examination of another dog was performed at a private veterinary clinic with submission of tissues to the UF CVM for histopathologic diagnosis. Tissues were fixed in 10% neutral buffered formalin, embedded in paraffin, and 5-μm sections were either stained with hematoxylin and eosin for histopathologic diagnosis or processed for immunohistochemistry as described below. Unfixed tissues were submitted for bacterial culture and also stored at −80° C.


Serological Tests for Canine Viral Respiratory Pathogens.


Paired acute and convalescent serum samples were submitted to the Animal Health Diagnostic Laboratory (AHDL) at the Cornell University College of Veterinary Medicine for serum neutralization assays against canine distemper virus, adenovirus type 2, and parainfluenza virus. Antibody titers were expressed as the last dilution of serum that inhibited viral infection of cell cultures. Seroconversion, defined as a ≥4-fold increase in antibody titer between the acute and convalescent sample, indicated viral infection. No seroconversions to these viral pathogens were detected.


Microbial Tests for Canine Bacterial Respiratory Pathogens.


Paired nasal swabs and postmortem tissues were submitted to the Diagnostic Clinical Microbiology/Parasitology/Serology Service at the UF CVM for bacterial isolation and identification. The samples were cultured on nonselective media as well as media selective for Bordetella species (Regan-Lowe; Remel) and Mycoplasma species (Remel). All cultures were held for 21 days before reporting no growth. Nasal swabs from some of the greyhounds were also submitted to the Department of Diagnostic Medicine/Pathobiology at the Kansas State University College of Veterinary Medicine for bacterial culture. Of 70 clinically diseased dogs tested, Bordetella bronchiseptica was isolated from the nasal cavity of 1 dog, while Mycoplasma spp. were recovered from the nasal cavity of 33 dogs. Pasteurella multocida was commonly recovered from the nasal cavity of dogs with purulent nasal discharges. Two of the dogs that died in the January 2004 outbreak had scant growth of Escherichia coli in the lungs postmortem, one dog had scant growth of E. coli and Streptococcus canis, and another had scant growth of Pseudomonas aeruginosa and a yeast. Neither Bordetella bronchiseptica nor Mycoplasma was isolated from the trachea or lungs of dogs that died.


Virus Isolation from Postmortem Tissues.


Frozen tissues were thawed and homogenized in 10 volumes of minimum essential medium (MEM) supplemented with 0.5% bovine serum albumin (BSA) and antibiotics. Solid debris was removed by centrifugation and supernatants were inoculated onto cultured cells or into 10-day old embryonated chicken eggs. Tissue homogenates from greyhounds that died were inoculated into diverse cell cultures that supported the replication of a broad range of viral pathogens. The cell cultures included Vero (African green monkey kidney epithelial cells, ATCC No. CCL-81), A-72 (canine tumor fibroblasts, CRL-1542), HRT-18 (human rectal epithelial cells, CRL-11663), MDCK (canine kidney epithelial cells, CCL-34), primary canine kidney epithelial cells (AHDL, Cornell University), primary canine lung epithelial cells (AHDL), and primary bovine testicular cells (AHDL). MDCK and HRT cells were cultured in MEM supplemented with 2.5 ug/mL TPCK-treated trypsin (Sigma); the remaining cell lines were cultured in MEM supplemented with 10% fetal calf serum and antibiotics. Cells were grown in 25 cm2 flasks at 37° C. in a humidified atmosphere containing 5% CO2. A control culture was inoculated with the supplemented MEM. The cultures were observed daily for morphologic changes and harvested at 5 days post inoculation. The harvested fluids and cells were clarified by centrifugation and inoculated onto fresh cells as described for the initial inoculation; two blind passages were performed. Hemagglutination activity in the clarified supernatants was determined using chicken or turkey red blood cells as described (Burleson et al., 1992; Kendal et al., 1982). For virus isolation in chicken embryos, 0.1 mL of tissue homogenate was inoculated into the allantoic sac and incubated for 48 hours at 35° C. After two blind passages, the hemagglutination activity in the allantoic fluids was determined as described (Burleson et al., 1992; Kendal et al., 1982).


RT-PCR, Nucleotide Sequencing, and Phylogenetic Analyses.


Total RNA was extracted from tissue culture supernatant or allantoic fluid using the RNeasy kit (Qiagen, Valencia, CA) according to manufacturer's instructions. The total RNA (10 ng) was reverse transcribed to cDNA using a one-step RT-PCR Kit (Qiagen, Valencia, CA) according to manufacturer's instructions. PCR amplification of the coding region of the 8 influenza viral genes in the cDNA was performed as previously described (Klimov et al., 1992a), using universal gene-specific primer sets. The resulting DNA amplicons were used as templates for automated sequencing on an Applied Biosystems 3100 automated DNA sequencer using cycle sequencing dye terminator chemistry (ABI). Nucleotide sequences were analyzed using the GCG Package©, Version 10.0 (Accelyrs) (Womble, 2000). The Phylogeny Inference Package© Version 3.5 was used to estimate phylogenies and calculate bootstrap values from the nucleotide sequences (Felsenstein, 1989). Phylogenetic trees were compared to those generated by neighbor-joining analysis with the Tamura-Nei gamma model implemented in the MEGA© program (Kumar et al., 2004) and confirmed by the PAUP© 4.0 Beta program (Sinauer Associates).


Experimental Inoculation of Dogs.


Four 6-month old specific pathogen-free beagles [(2 males and 2 females (Liberty Research)] were used. Physical examination and baseline blood tests including complete blood cell count/differential, serum chemistry panel, and urinalysis determined that the animals were healthy. They were housed together in a BSL 2-enhanced facility accredited by the Association for Assessment and Accreditation of Laboratory Animal Care. Baseline rectal temperatures were recorded twice daily for 7 days. The dogs were anesthetized by intravenous injection of propofol (Diprivan®, Zeneca Pharmaceuticals, 0.4 mg/kg body weight to effect) for intubation with endotracheal tubes. Each dog was inoculated with a total dose of 106.6 median tissue culture infectious doses (TCID50) of A/Canine/Florida/43/2004 (Canine/FL/04) (H3N8) virus with half the dose administered into the distal trachea through the endotracheal tube and the other half administered into the deep nasal passage through a catheter. Physical examinations and rectal temperature recordings were performed twice daily for 14 days post inoculation (p.i.). Blood samples (4 mL) were collected by jugular venipuncture on days 0, 3, 5, 7, 10, and 14 p.i. Nasal and oropharyngeal specimens were collected with polyester swabs (Fisher Scientific) from each dog on days 0 to 5, 7, 10, and 14 p.i. The swabs were placed in viral transport medium (Remel) and stored at −80° C. Two dogs (1 male and 1 female) were euthanatized by intravenous inoculation of Beuthanasia-D® solution (1 mL/5 kg body weight; Schering-Plough Animal Health Corp) on day 5 p.i. and the remaining 2 dogs on day 14 for postmortem examination. Tissues for histological analysis were processed as described. Tissues for virus culture were stored at −80° C. This study was approved by the University of Florida Institutional Animal Care and Use Committee.


Virus Shedding from Experimentally Inoculated Dogs.


Serial dilutions of lung homogenates and swab extracts, prepared by clarification of the swab transport media by centrifugation, were set up in MEM supplemented with 0.5% BSA and antibiotics. Plaque assays were performed as described (Burleson et al., 1992) using monolayers of MDCK cells in 6-well tissue culture plates. Inoculated cell monolayers were overlaid with supplemented MEM containing 0.8% agarose and 1.5 ug/mL of TPCK-trypsin. Cells were cultured for 72 hours at 37° C. in a humidified atmosphere containing 5% CO2 prior to fixation and staining with crystal violet. Virus concentration was expressed as plaque forming units (PFU) per gram of tissue or per swab.


Immunohistochemistry.


Deparaffinized and rehydrated 5 μm lung tissue sections from the greyhounds and beagles were mounted on Bond-Rite™ slides (Richard-Allan Scientific, Kalamazoo, MI) and subsequently treated with proteinase K (DakoCytomation, Carpenteria, CA) followed by peroxidase blocking reagent (Dako® EnVision™ Peroxidase Kit, Dako Corp.). The sections were incubated with 1:500 dilutions of monoclonal antibodies to canine distemper virus (VMRD, Inc.), canine adenovirus type 2 (VMRD, Inc.), canine parainfluenza virus (VMRD, Inc.), or influenza A H3 (Chemicon International, Inc.) for 2 hours at room temperature. Controls included incubation of the same sections with mouse IgG (1 mg/mL, Serotec, Inc.), and incubation of the monoclonal antibodies with normal canine lung sections. Following treatment with the primary antibodies, the sections were incubated with secondary immunoperoxidase and peroxidase substrate reagents (Dako® EnVision™ Peroxidase Kit, Dako Corp.) according to the manufacturer's instructions. The sections were counterstained with hematoxylin, treated with Clarifier #2 and Bluing Reagent (Richard-Allan Scientific, Kalamazoo, MI), dehydrated, and coverslips applied with Permount (ProSciTech).


Hemagglutination Inhibition (HI) Assay.


Serum samples were incubated with receptor destroying enzyme (RDE, Denka) (1 part serum: 3 parts RDE) for 16 hours at 37° C. prior to heat inactivation for 60 minutes at 56° C. Influenza A/Canine/FL/04 (H3N8) virus was grown in MDCK cells for 36-48 hr at 37° C. Virus culture supernatants were harvested, clarified by centrifugation, and stored at −80° C. The HI assay was performed as described previously (Kendal et al., 1982). Briefly, 4 hemagglutinating units of virus in 25 μl were added to an equal volume of serially diluted serum in microtiter wells and incubated at room temperature for 30 minutes. An equal volume of 0.5% v/v turkey erythrocytes was added and the hemagglutination titers were estimated visually after 30 minutes. The endpoint HI titer was defined as the last dilution of serum that completely inhibited hemagglutination. Seroconversion was defined as ≥4-fold increase in HI titer between paired acute and convalescent samples. Seropositivity of a single sample was defined as a HI antibody titer ≥1:32.


Microneutralization (MN) Assay.


Neutralizing serum antibody responses to A/Canine/FL/04 (H3N8) were detected by a MN assay as described previously (Rowe et al., 1999) except that canine sera were RDE-treated as described above prior to the assay. The endpoint titer was defined as the highest dilution of serum that gave 50% neutralization of 100 TCID50 of virus. Seroconversion was defined as ≥4-fold increase in MN titer between paired acute and convalescent samples. Seropositivity of a single sample was defined as a MN titer ≥1:80.


Following are examples which illustrate procedures for practicing the invention. These examples should not be construed as limiting. All percentages are by weight and all solvent mixture proportions are by volume unless otherwise noted.


Example 1

In January 2004, an outbreak of respiratory disease occurred in 22 racing greyhounds housed in 2 kennels at a Florida track and the local farm that supplied dogs to these kennels. There were approximately 60 dogs in each kennel building and 300 dogs at the farm. The outbreak occurred over a 6-day period after which no new cases were identified. Fourteen of the 22 dogs had fevers of 39.5 to 41.5° C., a soft, gagging cough for 10 to 14 days, and eventual recovery. Of the remaining 8 dogs, 6 apparently healthy dogs died unexpectedly with hemorrhage from the mouth and nose. Two other dogs were euthanatized within 24 hours of onset of hemorrhage from the mouth and nose due to rapid deterioration. Both of these dogs had fevers of 41° C. Four of the 8 deaths occurred in the kennel buildings and 4 occurred at the farm. Fifty percent of the deaths occurred on day 3 of the outbreak. The 22 dogs ranged in age from 17 months to 4 years, but 73% were 17 to 33 months old.


Two clinical syndromes were evident: a milder illness characterized by initial fever and then cough for 10-14 days (14 dogs) with subsequent recovery, or a peracute death associated with hemorrhage in the respiratory tract (8 dogs for a mortality rate of 36%). Postmortem examinations were performed on 6 of the 8 fatal cases. All dogs had extensive hemorrhage in the lungs, mediastinum, and pleural cavity. Histological examination of the respiratory tract revealed that in addition to pulmonary hemorrhage, all dogs had tracheitis, bronchitis, bronchiolitis, and suppurative bronchopneumonia (FIGS. 3A and 3B). The epithelial lining and airway lumens in these tissues were infiltrated by neutrophils and macrophages. Lung homogenates prepared from these dogs were inoculated into a variety of monkey, human, bovine, and canine cell lines for virus culture. The lung homogenate from one dog caused cytopathic effects in Madin-Darby canine kidney epithelial cells (MDCK) cultured in the presence of trypsin, and the cell culture supernatant agglutinated chicken red blood cells. Preliminary evidence of an influenza type A virus was provided by a commercial ELISA for detection of the nucleoprotein of influenza A and B viruses, and by PCR analysis using primers specific for the matrix gene of influenza A viruses. In addition, the hemagglutinating activity was inhibited by reference antisera to the equine influenza A H3 subtype, but not by antisera specific for human influenza A subtypes H1-H11 and H13 (Table 3). To characterize the molecular properties of the virus, we determined the nucleotide sequences of the 8 RNA segments of the viral genome. Sequence comparisons with known influenza virus genes and phylogenetic analyses indicated that the 8 genes of the canine isolate were most similar to those from contemporary equine influenza A (H3N8) viruses, with which they shared ≥96-97% sequence identity (FIG. 1A, Table 4). In contrast, representative genes from avian, swine, and human influenza A isolates had ≤94% identity with the canine isolate (Table 4). These data identified the canine isolate A/Canine/Florida/43/2004 (Canine/FL/04) as an influenza A H3N8 virus closely related to contemporary lineages of equine influenza viruses. Since all genes of the canine isolate were of equine influenza virus origin, we concluded that the entire genome of an equine influenza virus had been transmitted to the dog.


Example 2

To investigate the role of the Canine/FL/04 virus in the clinical and pathological observations in the greyhounds, we performed immunohistochemical staining (IHC) on lung tissues using a monoclonal antibody to influenza A H3. Viral H3 antigen was consistently detected in the cytoplasm of bronchial and bronchiolar epithelial cells, bronchial gland epithelial cells, and macrophages in airway lumens and alveolar spaces (FIG. 2A). These data support a diagnosis of pulmonary infection with influenza virus of the H3 subtype in multiple dogs.


Example 3

To determine involvement of a Canine/FL/04-like virus in the etiology of the respiratory disease outbreak, we analyzed paired acute and convalescent sera from 11 sick dogs and 16 asymptomatic contacts by hemagglutination inhibition (HI) and microneutralization (MN). Seroconversion, defined as a ≥4-fold rise in antibody titer to Canine/FL/04 from the acute to convalescent phase, occurred in 8 of 11 (73%) sick dogs in both assays (Table 1). Seroconversion occurred in 6 of 16 (38%) asymptomatic contacts in the HI assay, while 8 of 16 (50%) seroconverted in the MN assay (Table 1). The seroconversion data demonstrated infection of the dogs with a Canine/FL/04-like virus which coincided temporally with the onset of respiratory disease in most animals.


Single serum samples were collected 3 months after the outbreak from an additional 46 asymptomatic dogs housed with the sick dogs. Of these, 43 (93%) were seropositive in both assays. For the total population of 73 dogs tested, 93% were seropositive in both assays, including 82% (9/11) of the sick dogs and 95% (59/62) of the healthy contacts. The high seroprevalence in dogs with no history of respiratory disease indicates that most infections with canine influenza virus are subclinical and suggest efficient spread of the virus among dogs. It is not known if subclinical infections contribute to the spread of the virus.


Example 4

To better understand the capacity of the Canine/FL/04 virus to infect dogs, four 6-month old purpose-bred beagles were each inoculated with 106.6 median tissue culture infectious doses (TCID50) by the intratracheal and intranasal routes. All dogs developed a fever (rectal temperature ≥39° C.) for the first 2 days postinoculation (p.i.), but none exhibited respiratory symptoms such as cough or nasal discharge over a 14 day observation period. Virus shedding was examined by quantification of virus in nasal and oropharyngeal swabs. Only 2 of the 4 dogs shed detectable amounts of virus. One dog shed virus on days 1 and 2 p.i. (1.0-2.5 log10 PFU per swab), whereas the other dog shed virus for 4 consecutive days after inoculation (1.4-4.5 log10 PFU per swab). Postmortem examination of 2 dogs on day 5 p.i. revealed necrotizing and hyperplastic tracheitis, bronchitis, and bronchiolitis similar to that found in the spontaneous disease in greyhounds, but there was no pulmonary hemorrhage or bronchopneumonia. Viral H3 antigen was detected in the cytoplasm of epithelial cells of bronchi, bronchioles, and bronchial glands by IHC (FIG. 2B). Infectious virus was recovered from the lung tissue of one of the dogs. Postmortem examination of the remaining 2 dogs on day 14 p.i. showed minimal histological changes in respiratory tissues, no viral H3 antigen by IHC, and no recovery of virus from lung homogenates. Seroconversion in these latter 2 dogs was detected in MN assays by day 7 p.i., with a further 2 to 3-fold increase in antibody titers by day 14. These results established the susceptibility of dogs to infection with Canine/FL/04, as evidenced by the febrile response, presence of viral antigen and infectious virus in the lung parenchyma, histopathological findings typical for influenza, and seroconversion. The failure to reproduce severe disease and death in the experimentally inoculated beagles is not surprising since a large proportion of the naturally infected greyhounds were asymptomatic.


Example 5

To investigate whether a Canine/FL/04-like influenza virus had circulated among greyhound populations in Florida prior to the January 2004 outbreak, archival sera from 65 racing greyhounds were tested for the presence of antibodies to Canine/FL/04 using the HI and MN assays. There were no detectable antibodies in 33 dogs sampled from 1996 to 1999. Of 32 dogs sampled between 2000 and 2003, 9 were seropositive in both assays—1 in 2000, 2 in 2002, and 6 in 2003 (Table 5). The seropositive dogs were located at Florida tracks involved in outbreaks of respiratory disease of unknown etiology from 1999 to 2003, suggesting that a Canine/FL/04-like virus may have been the causative agent of those outbreaks. To investigate this possibility further, we examined archival tissues from greyhounds that died from hemorrhagic bronchopneumonia in March 2003. Lung homogenates inoculated into MDCK cells and chicken embryos from one dog yielded H3N8 influenza virus, termed A/Canine/Florida/242/2003 (Canine/FL/03). Sequence analysis of the complete genome of Canine/FL/03 revealed >99% identity to Canine/FL/04 (Table 4), indicating that Canine/FL/04-like viruses had infected greyhounds prior to 2004.


Example 6

From June to August 2004, respiratory disease outbreaks occurred in thousands of racing greyhounds at 14 tracks in Florida, Texas, Alabama, Arkansas, West Virginia, and Kansas.


Officials at some of these tracks estimated that at least 80% of their dog population had clinical disease. Most of the dogs had clinical signs of fever (≥39° C.) and cough similar to the dogs in the January 2004 outbreak, but many dogs also had a mucopurulent nasal discharge. Multiple deaths were reported but an accurate mortality rate could not be determined.


We collected paired acute and convalescent sera from 94 dogs located at 4 Florida tracks: 56% of these dogs had ≥4-fold rises in antibody titers to Canine/FL/04, and 100% were seropositive (Table 6). Convalescent sera from 29 dogs in West Virginia and Kansas also had antibodies to Canine/FL/04. We isolated influenza A (H3N8) virus from the lungs of a greyhound that died of hemorrhagic bronchopneumonia at a track in Texas. Sequence analysis of the entire genome of this isolate, named A/Canine/Texas/1/2004 (Canine/TX/04), revealed ≥99% identity to Canine/FL/04 (Table 4). The isolation of three closely related influenza viruses from fatal canine cases over a 13-month period and from different geographic locations, together with the substantial serological evidence of widespread infection among racing greyhounds, suggested sustained circulation of a Canine/FL/04-like virus in the dog population.


Phylogenetic analysis of the HA genes of Canine/FL/03, Canine/FL/04, and Canine/TX/04 showed that they constitute a monophyletic group with robust bootstrap support that was clearly distinct from contemporary H3 genes of equine viruses isolated in 2002 and 2003 (FIG. 1B). Phylogentic analysis and pairwise nucleotide sequence comparisons of the other 7 genomic segments supported the segregation of the canine genes as a distinct sub-lineage most closely related to the equine virus lineage (data not shown, and Table 4). The clustering of the canine influenza genes as a monophyletic group separate from equine influenza is also supported by the presence of 4 signature amino acid changes in the HA (Table 2). Together with the serological results from 2003 and 2004, these data are consistent with a single virus transmission event from horses to dogs with subsequent horizontal spread of the virus in the greyhound population. However, repeated introductions of this unique lineage of influenza virus from an unidentified reservoir species can not be formally excluded, unlikely as it may be.


The viral HA is a critical determinant of host species specificity of influenza virus (Suzuki et al., 2000). To identify residues within HA that may be associated with adaptation to the canine host, we compared the deduced amino acid sequence of canine HAs to those of contemporary equine viruses. Four amino acid changes differentiate the equine and canine mature HA consensus amino acid sequences: N83S, W222L, I328T, and N483T (see Table 2). The canine viruses have an amino acid deletion when compared to the consensus equine sequences. Therefore, amino acid position 7 in the HA equine sequence is position 6 in the HA canine sequence, amino acid position 29 in the HA equine sequence is position 28 in the HA canine sequence, amino acid position 83 in the HA equine sequence is position 82 in the HA canine sequence, etc. Thus, the four substituted amino acids are at position 82, 221, 327, and 482 of the amino acid sequence shown in SEQ ID NO: 33 and SEQ ID NO: 34. The substitution of serine for asparagine at consensus sequence position 83 is a change of unknown functional significance since various polar residues are found in H3 molecules from other species. The strictly conserved isoleucine at consensus sequence position 328 near the cleavage site of the H3 HA has been replaced by threonine. The pivotal role of HA cleavage by host proteases in pathogenesis suggests that this change merits further study. The substitution of leucine for tryptophan at consensus sequence position 222 is quite remarkable because it represents a non-conservative change adjacent to the sialic acid binding pocket which could modulate receptor function (Weis et al., 1988). Interestingly, leucine at position 222 is not unique to canine H3 HA since it is typically found in the H4, H8, H9, and H12 HA subtypes (Nobusawa et al., 1991; Kovacova et al., 2002). The leucine substitution may be more compatible with virus specificity for mammalian hosts since infections of swine with subtype H4 (Karasin et al., 2000) and humans and swine with subtype H9 (Peiris et al., 1999) viruses have been reported. The substitution of asparagine with threonine at consensus sequence position 483 resulted in the loss of a glycosylation site in the HA2 subunit that is conserved in all HA subtypes (Wagner et al., 2002). Although the importance of these amino acid changes in the HA for adaptation of an equine virus to dogs remains to be determined, similar amino acid changes have been observed previously in association with interspecies transfer (Vines et al., 1998; Matrosovich et al., 2000). Amino acid differences between other influenza viral proteins of the invention and equine consensus sequence are shown in Tables 19 to 25.


The source of the equine influenza virus that initially infected racing greyhounds remains speculative. Kennels at greyhound racetracks are not located near horses or horse racetracks, suggesting that contact between greyhounds and shedding horses is not a sufficient explanation for the multiple outbreaks in different states in 2004. A potential source of exposure to the equine virus is the feeding of horsemeat to greyhounds, whose diet is supplemented with raw meat supplied by packing houses that render carcasses, including horses which could carry influenza. Precedents for this mode of infection include reports of interspecies transmission of H5N1 avian influenza virus to pigs and zoo felids fed infected chicken carcasses (Webster, 1998; Keawcharoen et al., 2004; Kuiken et al., 2004). Although this is a plausible route for the initial introduction of equine influenza into dogs, it does not explain the recent multiple influenza outbreaks in thousands of dogs in different states. Our experimental inoculation study demonstrated the presence of virus in the nasal passages and oropharynx of dogs, albeit at modest titers. Nevertheless, these results indicate that shedding is possible, and that dog-to-dog transmission of virus by large droplet aerosols, fomites, or direct mucosal contact could play a role in the epizootiology of the disease.


The interspecies transfer of a whole mammalian influenza virus to an unrelated mammal species is a rare event. Previous studies have provided limited serological or virological evidence, but not both, of transient infection of dogs with human influenza A (H3N2) viruses (Nikitin et al., 1972, Kilbourne, et al., 1975; Chang et al., 1976; Houser et al., 1980). However, there was no evidence of sustained circulation in the canine host. Although direct transfer of swine influenza viruses from pigs to people is well-documented (Dacso et al., 1984; Kimura et al., 1998; Patriarca et al., 1984; Top et al., 1977), there is no evidence for adaptation of the swine viruses in human hosts. In this report, we provide virological, serological, and molecular evidence for interspecies transmission of an entire equine influenza A (H3N8) virus to another mammalian species, the dog. Unique amino acid substitutions in the canine virus HA, coupled with serological confirmation of infection of dogs in multiple states in the U.S., suggest adaptation of the virus to the canine host. Since dogs are a primary companion animal for humans, these findings have implications for public health; dogs may provide a new source for transmission of novel influenza A viruses to humans.









TABLE 1







Antibody response to A/Canine/Florida/43/2004 (H3N8).










Sick Dogs (11)a
Healthy Contacts (16)b











Response
HIc
SNd
HI
SN














Seroconversion (%)e
73
73
38
50


Seropositive (%)f
82
82
100
100


Geometric mean titerg
329
424
268
431






aNumber of dogs with clinical signs of disease.




bNumber of asymptomatic dogs housed in contact with clinically diseased dogs.




cHemagglutination-inhibition (HI) assay using A/Canine/Florida/43/2004 virus.




dMicroneutralization (MN) assay using A/Canine/Florida/43/2004 virus.




ePercentage of dogs with at least a 4-fold increase in antibody titer in paired acute and convalescent sera.




fPercentage of dogs with a positive antibody titer (HI titer ≥32: MN titer ≥80) in the convalescent sera.




gGeometric mean antibody titer for the convalescent sera.














TABLE 2







Amino acid differences between the canine and


equine H3 hemagglutinins.











Equine H3
Can/
Can/
Can/



consensus
FL/03
FL/04
TX/04
Potential functional significance





G7*
D



D also found in duck






and human H3 HA


I29

M
M
I is conserved in H3 HAs from all






species


N83
S
S
S
Various polar amino acids present






at this position in H3 HAs of other






species


S92

N

N is present in some duck H3 HAs


L118


V
L is conserved in all H3 HAs


W222
L
L
L
W is conserved in most H3 HAs of






all species; located near the






receptor binding site


A272
V
A
V
V is present in some recent equine






isolates


I328
T
T
T
T is strictly conserved in all avian,






swine or humans H3 HAs


N483
T
T
T
N occurs in all H3 and other HA






subtypes. Replacement results in






loss of a glycosylation site.


K541

R

Basic amino acid conservative






change





*Amino acid residue (single letter code) and position in the mature H3 HA. The amino acid code is: A = alanine, D = aspartic acid, G = glycine, I = isoleucine, K = lysine, L = leucine, M = methionine, N = asparagine, R = arginine, S = serine, T = threonine, V = valine, W = tryptophan.


† Denotes no change from the consensus equine H3 HAs.













TABLE 3







Hemagglutination inhibition of a virus isolate by


reference antisera to different HA subtypes.











Reference Antisera
HA Specificity
HI titera















Puerto Rico/8/34
H1
5



Swine/Iowa15/30
H1
5



Singapore/01/57
H2
5



Shanghai/11/87

H3b

5



Equine/Miami/1/63
H3
160



Duck/Czechoslovakia/56
H4
5



Tern/South Africa/61
H5
5



Turkey/Massachussetts/65
H6
5



Fowl Plague/Dutch/27
H7
5



Fowl Plague/Rostock/34
H7
5



Equine/Prague/1/56
H7
5



Turkey/Ontario/6118/68
H8
5



Quail/Hong Kong/G1/97

H9b

5



Chicken/Hong Kong/G9/97

H9b

5



Chicken/Germany/49
 H10
5



Duck/England/56
 H11
5



Gull/Maryland/704/77
 H13
5



Normal sheep serum

5



Normal ferret serum

5








aHemagglutination inhibition titer to virus isolate from dog #43.





bPolyclonal antisera were produced in ferrets, whereas all other antisera were produced in sheep or goats.














TABLE 4







Sequence homology of A/Canine/Florida/43/2004 (H3N8) genes to equine, avian,


swine, and human strains of influenza A.











Gene
Equine
Avian
Swine
Human





PB2
96.9 (98.7)a
88.6 (96.8)
87.9 (96.8)
86.2 (96.4)


DQ124147
Eq/Kentucky/2/8
Mall/Alberta/98/85
Sw/Ontario/
PR/8/34



M73526
AY633315
01911-1/99
(HK/213/03)





AF285892
AF389115






(AY576381)


PB1
97.1 (98.8)
83.9 (97.1)
83.9 (97.1)
83.9 (97.1)


DQ124148
Eq/Tennessee/5/86
Ck/BritishColumbia/04
Sw/Korea/S109/04
WSN/33



M25929
(Gull/Md/704/77)
(Sw/Saskatch/
(Sing/1/57)




AY61675 (M25933)
18789/02)
J02178





AY790287
(M25924)





(AY619955)



PA
96.3 (97.5)
87.0 (94.3)
84.3 (94.6)
83.8 (93.4)


DQ124149
M26082
Ck/Chile/4591/02
Sw/Hong Kong/
Taiwan/2/70



Eq/Tennesee/5/86
(Ostrich/SA/08103/95)
126/02
(Viet Nam/




AY303660(AF508662)
M26081
1203/04)






AY210199






(AY818132)


HA (H3)
97.4 (97.1)
80.7 (89.0)
80.0 (87.7)
81.8 (87.9)


DQ124190
Eq/FL/1/93
Dk/Norway/1/03
Sw/Ontario/42729a/01
HK/1/68



L39916
AJ841293
AY619977
AF348176


NP
96.6 (97.9)
87.9 (95.1)
85.4 (93.5)
84.7 (93.0)


DQ124150
Eq/Tennesee/5/86
Ck/Chile/176822/02
Sw/Ontario/42729a/01
HK/1073/99



M30758
AY303658
(Sw/Fujian/1/2003)
(Hong Kong/





AY619974
538/97)





(AY747611)
AF255742






(AF255751)


NA (N8)
96.8 (97.0)
84.0 (85.2)
nab
nab


DQ124151
Eq/Tennesee/5/86
Dk/NJ/2000





L06583
L06583




M
97.9 (95.7)
94.1 (94.0)
93.7 (93.5)
91.2 (95.4)


DQ124152
Eq/Tennesee/5/86
Tky/Mn/833/80
Sw/Saskatchewan/
WSN/33



(Eq/Kentucky/92)
AF001683
18789/02
(Hong Kong/



M63529

M63527
1073/99)



(AF001683)


J02177






(AJ278646)


NS
97.5 (95.7)
92.0 (90.4)
91.1 (89.1)
91.4 (90.0)


DQ124153
Eq/Tn/5/86
Mal/NY/6750/78
Sw/China/8/78
Brevig



(Eq/Kentucky/92)
M80945
(Sw/Korea/s452/04)
Mission/1/18



M80973

M80968 (AY790309)
AF333238



(AF001671)






aPercent nucleotide and amino acid (in parentheses) sequence identity of A/Canine/Florida/43/2004 (H3N8) genes to the most homologous gene of influenza virus virus isolates from the species, followed by their Genbank sequence database accession numbers.




bNot applicable: N8 neuraminidase was never reported in human or swine viruses.














TABLE 5







Antibody titers to A/canine/Florida/43/2004 (H3N8) in greyhound


serum collected from 1996 to 2003.









Yeara














1996
1997
1998
2000
2002
2003
















No. of dogs tested
8
6
19
4
6
22


No. of seropositive dogs
0
0
0
1
2
6


Antibody titersb



512
232, 524
280-2242






aThe year of serum sample collection from racing greyhounds in Florida.




bMicroneutralization assay antibody titers for seropositive dogs, including the range for the six 2003 seropositive dogs.














TABLE 6







Antibody response to A/canine/Florida/43/2004 (H3N8) in racing


greyhounds at 4 Florida tracks in June 2004.











Response
Track A
Track B
Track C
Track D














Number of dogs testeda
37
10
22
25


Seroconversion (%)b
46
90
100
64


Seropositive (%)c
100
100
100
100


Geometric mean titerd
401
512
290
446






aNumber of clinically diseased dogs tested by HI using A/canine/Florida/43/2004 (H3N8).




bPercentage of dogs with ≥ 4-fold increase in antibody titer between acute and convalescent sera.




cPercentage of dogs with a positive antibody titer (HI titer > 16) in the convalescent sera.




dGeometric mean antibody titer for the convalescent sera.







Materials and Methods for Examples 7-11

Canine Tissues


Postmortem examinations were performed by the Anatomic Pathology Service at the University of Florida College of Veterinary Medicine on 6 mixed breed dogs that died in April/May 2005 during an influenza outbreak in a shelter facility in northeast Florida, and on a pet Yorkshire Terrier dog that died in May 2005 during an influenza outbreak in a veterinary clinic in southeast Florida. Tissues were fixed in 10% neutral buffered formalin, embedded in paraffin, and 5-μm sections were stained with hematoxylin and eosin for histopathologic diagnosis. Unfixed tissues were stored at −80° C. pending virological analyses.


RNA Extraction from Canine Tissue Samples


Frozen lung tissues from each of the 7 dogs were thawed and homogenized in minimum essential medium (MEM) supplemented with 0.5% bovine serum albumin (BSA) and antibiotics (gentamycin and ciprofloxacin) using a disposable tissue grinder (Kendall, Lifeline Medical Inc., Danbury, CT). Total RNA was extracted using a commercial kit (RNeasy® Mini Kit, QIAGEN Inc., Valencia, CA) according to manufacturer's instructions and eluted in a final volume of 60 μL of buffer. Total RNA was also extracted from lung tissue collected from dogs without respiratory disease.


Real-time RT-PCR


A single-step quantitative real-time RT-PCR was performed on total RNA extracted from the canine tissue samples using the QuantiTect® Probe RT-PCR Kit containing ROX as a passive reference dye (QIAGEN Inc., Valencia, CA). Briefly, 2 primer-probe sets were used for detection of influenza A sequences in each sample (Table 7). One primer-probe set was selective for canine hemagglutinin (H3) gene sequences. The other primer-probe set targeted a highly conserved region of the matrix (M) gene of type A influenza virus. For each real-time RT-PCR reaction, 5 μL of extracted total RNA were added to a reaction mixture containing 12.5 μL of 2× QuantiTech® Probe RT-PCR Master Mix, 0.25 μL of QuantiTech® RT Mix, forward and reverse primers (0.4 μM final concentration for each), probe (0.1 μM final concentration) and RNase-free water in a final volume of 25 pt. The TaqMan® Ribosomal RNA Control Reagents (Applied Biosystems, Foster City, CA) were used according to manufacturer's instructions for detection of 18S rRNA as an endogenous internal control for the presence of RNA extracted from the canine tissue samples.


Quantitative one-step real-time RT-PCR was performed on the reaction mixtures in a Mx3000P® QPCR System (Stratagene, La Jolla, CA). Cycling conditions included a reverse transcription step at 50° C. for 30 minutes, an initial denaturation step at 95° C. for 15 minutes to activate the HotStarTaq® DNA polymerase, and amplification for 40 cycles. Each amplification cycle included denaturation at 94° C. for 15 seconds followed by annealing/extension at 60° C. for 1 minute. The FAM (emission wavelength 518 nm) and VIC (emission wavelength 554 nm) fluorescent signals were recorded at the end of each cycle. The threshold cycle (Ct) was determined by setting the threshold fluorescence (dR) at 1000 in each individual experiment. The Mx3000P® version 2.0 software program (Stratagene, La. Jolla, CA) was used for data acquisition and analysis. Samples were considered positive for influenza A virus when the threshold cycle (Ct) for the H3 or M gene was 3 units smaller than the Ct for lung tissues from dogs without respiratory disease. The positive control consisted of amplification of RNA extracted from A/canine/FL/242/03 (H3N8) virus.


Virus Isolation in MDCK Cells


Frozen lung tissues from each of the 7 dogs were thawed and homogenized in 10 volumes of Dulbecco's Modified Eagle Medium (DMEM) supplemented with 0.5% (BSA) and antibiotics (gentamycin and ciprofloxacin). Solid debris was removed by centrifugation and supernatants were inoculated onto Madin-Darby canine kidney (MDCK) cells cultured in DMEM supplemented with 1 μg/mL TPCK-treated trypsin (Sigma-Aldrich Corp., St. Louis, MO) and antibiotics (gentamycin and ciprofloxacin). Cells were grown in 25 cm2 flasks at 37° C. in a humidified atmosphere containing 5% CO2. The cultures were observed daily for morphologic changes and harvested at 5 days post inoculation. The harvested cultures were clarified by centrifugation and the supernatants inoculated onto fresh MDCK cells as described for the initial inoculation; two additional passages were performed for samples that did not show evidence of influenza virus by hemagglutination or RT-PCR. Hemagglutination activity in the clarified supernatants was determined using 0.5% turkey red blood cells as previously described (Burleson, F. et al., 1992; Kendal, P. et al., 1982). RT-PCR was performed as described below.


Virus Isolation in Embryonated Chicken Eggs


Homogenates were prepared from frozen lung tissues as described above for inoculation of MDCK cells. The homogenates (0.2 mL) were inoculated into the allantoic sac of 10-day old embryonated chicken eggs. After 48 hours of incubation at 35° C., the eggs were chilled at 4° C. overnight before harvesting the allantoic fluid. Hemagglutination activity in the clarified supernatants was determined using 0.5% turkey red blood cells as previously described (Burleson, F. et al., 1992; Kendal, P. et al., 1982). RT-PCR was performed as described below. Two additional passages in embryonated eggs were performed for samples that did not show evidence of influenza virus after the initial inoculation.


RT-PCR, Nucleotide Sequencing, and Phylogenetic Analyses


Viral RNA was extracted from MDCK supernatant or allantoic fluid using the QIAamp® Viral RNA Mini Kit (QIAGEN Inc., Valencia, CA) according to manufacturer's instructions. The viral RNA was reverse transcribed to cDNA using the QIAGEN® OneStep RT-PCR Kit (QIAGEN Inc., Valencia, CA) according to manufacturer's instructions. PCR amplification of the coding region of the 8 influenza viral genes in the cDNA was performed as previously described (Klimov, A. et al., 1992b), using universal gene-specific primer sets (primer sequences available on request). The resulting DNA amplicons were used as templates for automated sequencing in the ABI PRISM® 3100 automated DNA sequencer using cycle sequencing dye terminator chemistry (Applied Biosystems, Foster City, CA). Nucleotide sequences were analyzed using the Lasergene 6 Package® (DNASTAR, Inc., Madison, WI). The PHYLIP Version 3.5© software program was used to estimate phylogenies and calculate bootstrap values from the nucleotide sequences (Felsenstein, J., 1989). Phylogenetic trees were compared to those generated by neighbor-joining analysis with the Tamura-Nei model implemented in the MEGA© program (Kumar, S. et al., 2004) and confirmed by the PAUP© 4.0 Beta program (Sinauer Associates, Inc., Sunderland, MA).


Hemagglutination Inhibition (HI) Assay


Serum samples were incubated with receptor destroying enzyme (RDE, DENKA SEIKEN Co., Ltd., Tokyo, Japan) (1 part serum: 3 parts RDE) for 16 hours at 37° C. prior to heat inactivation for 30 minutes at 56° C. Influenza A/Canine/Jacksonville/05 (H3N8) virus was grown in MDCK cells for 72 hrs at 37° C. in 5% CO2. Virus culture supernatants were harvested, clarified by centrifugation, and stored at −80° C. All other viruses used in the HI assay were grown in 10-day old embryonated chicken eggs from which allantoic fluid was collected and stored at −80° C. The HI assay was performed as described previously (Kendal, P. et al., 1982). Briefly, 4 hemagglutinating units of virus in 25 μl were added to an equal volume of serially diluted serum in 96-well plastic plates and incubated at room temperature for 30 minutes. An equal volume of 0.5% turkey erythrocytes was added and the hemagglutination titers were estimated visually after 30 minutes. The endpoint HI titer was defined as the last dilution of serum that completely inhibited hemagglutination.


Example 7-Clinical Cases

In April and May 2005, a previously described (Crawford, P. C. et al., 2005) respiratory disease outbreak occurred in dogs housed in a shelter facility in northeast Florida. The outbreak involved at least 58 dogs ranging in age from 3 months to 9 years, and included purebred dogs as well as mixed breeds. The most common clinical signs were purulent nasal discharge and a cough for 7 to 21 days. Of the 43 dogs that had clinical disease for ≥7 days, 41 had HI antibody titers to canine/FL/04 (H3N8) ranging from 32 to >1024. At least 10 dogs progressed to pneumonia, of which 6 were euthanized. These 6 mixed breed dogs included 3 males and 3 females ranging in age from 4 months to 3 years. The duration of clinical signs ranged from 2 to 10 days at the time of euthanasia. On postmortem examination, these dogs had pulmonary congestion and edema. Histological examination of the respiratory tract revealed rhinitis, tracheitis, bronchitis, bronchiolitis, and suppurative bronchopneumonia. There was epithelial cell necrosis and erosion in the trachea, bronchi, bronchioles, and bronchial glands. The respiratory tissues were infiltrated by neutrophils and macrophages.


In May 2005, a respiratory disease outbreak occurred in 40 pet dogs at a veterinary clinic in southeast Florida. The most common clinical signs were purulent nasal discharge and a cough for 10 to 30 days. Of the 40 dogs, 17 were seropositive for canine/FL/04 (H3N8) with HI antibody tiers ranging from 32 to >1024. Seroconversion occurred in 10 dogs for which paired acute and convalescent sera were available. Three dogs progressed to pneumonia. One of these dogs, a 9-year old male Yorkshire Terrier, died 3 days after onset of clinical signs. This dog had tracheobronchitis, pulmonary edema and congestion, and severe bronchopneumonia. Similar to the 6 shelter dogs, there was epithelial cell necrosis and erosion of the airways and neutrophilic infiltrates in the tissues.


Example 8-Real-Time RT-PCR and Viral Isolation

Lung tissues from the 7 dogs were analyzed by quantitative real-time RT-PCR assays that detect the M gene of influenza type A and the H3 gene of canine H3N8 influenza A virus. The lungs from all 7 dogs were positive for both the influenza A M gene and the canine influenza H3 gene (Table 8). After 3 passages in MDCK cells, influenza A subtype H3N8 virus was isolated from the lungs of a shelter dog that died after 3 days of pneumonia. This virus was named A/canine/Jacksonville/05 (H3N8) (canine/Jax/05). After 2 passages in embryonated chicken eggs, influenza A subtype H3N8 virus was recovered from the lungs of the pet dog that also died after 3 days of pneumonia. This virus was named A/canine/Miami//05 (H3N8) (canine/Miami/05).


Example 9—Genetic Analyses of the Canine Influenza a H3N8 Isolates

Sequence analyses of canine/Jax/05 and canine/Miami/05 revealed that their hemagglutinin (HA) genes were 98% identical to the canine/FL/04, canine/TX/04, and canine/Iowa/05 isolates recovered from the lungs of racing greyhounds that died of pneumonia during influenza outbreaks at tracks in 2004 and 2005 (Crawford, P. C. et al., 2005; Yoon K-Y. et al., 2005). In addition, the HA genes of canine/Jax/05 and canine/Miami/05 were 98% identical to contemporary equine influenza viruses isolated after the year 2000. Phylogenetic comparisons of the HA genes showed that the canine/Jax/05 and canine/Miami/05 viruses were clustered with the canine/FL/04, canine/TX/04, and canine/Iowa/05 greyhound isolates and contemporary equine isolates, forming a distinct group from the older equine viruses isolated in the early 1990's (FIG. 4). Furthermore, the canine/Jax/05, canine/Miami/05, and canine/Iowa/05 isolates were more closely related to canine/Tx/04 than to either canine/FL/04 or canine/FL/03. The 2005 isolates formed a subgroup that appears to branch off from the earlier 2003 and 2004 canine viruses with differences at approximately 10 parsimony-informative sites. These differences support the hypothesis that canine influenza virus is being transmitted horizontally from dog-to-dog as opposed to being reintroduced periodically from an outside source. The accumulation of mutations from 2003 to 2005 illustrates the ongoing process of adaptation that the virus must undergo after being transmitted to a new host, as is expected to have happened for the canine influenza viruses.


Example 10-Amino Acid Analyses of the Canine Influenza a H3N8 Isolates

There were conserved amino acid substitutions in all 6 canine isolates that differentiated them from contemporary equine influenza viruses (Table 9). These conserved substitutions were 115M, N83S, W222L, I328T, and N483T. Phylogenetic comparisons of the mature HA protein showed that the canine/Jax/05, canine/Miami/05, and canine/Iowa/05 viruses formed a subgroup with the canine/TX/04 isolate (FIG. 4). There were 3 amino acid changes (L118V, K261N, and G479E) that differentiated this subgroup from the other canine viruses (Table 9). There were 2 amino acid changes (F79L and G218E) that differentiated the 2005 isolates from their canine/TX/04 root. Furthermore, the 2005 isolates from non-greyhound dogs, canine/Jax/05 and canine/Miami/05, differed from the canine/Iowa/05 greyhound isolate by one amino acid change, R492K. Finally, canine/Jax/05 differed from canine/Miami/05 at a single amino acid, S107P. In all other H3N8 equine and canine viruses, S is conserved at position 107 except for A/Equine/Jilin/1/89 which has a T (Guo Y. et al., 1992).


Example 11-Antigenic Analyses of the Canine Influenza a H3N8 Isolates

Hemagglutination inhibition (HI) tests were performed using an antigen panel of older and contemporary equine influenza viruses and the canine influenza viruses, and serum collected in 2005 from horses and dogs that had been infected with influenza virus (Table 10). Serum from ferrets immunized against canine/FL/04 was also included in the analyses. The HI antibody titers in equine serum were 8 to 16-fold higher when tested with contemporary equine viruses compared to older isolates, but decreased by at least 4-fold when tested with the canine viruses. The canine serum was nonreactive with the older equine viruses, but the antibody titers increased 4-fold when tested with contemporary equine isolates and canine isolates. This was also observed for the serum from ferrets immunized against canine influenza virus. These seroreactivity patterns demonstrated the antigenic similarity between the canine influenza viruses and contemporary equine influenza viruses and were consistent with the phylogenetic analyses. The antibody titers in equine, canine, and ferret sera to the canine/Miami/05 isolate were similar to those for the 2003 and 2004 canine isolates. However, the titers were 2 to 4-fold lower for the canine/Jax/05 isolate. This suggests that canine/Jax/05 is antigenically distinct from the other canine isolates, which may in part be related to the single amino acid change at position 107 in the mature HA.









TABLE 7







Primers and probes for quantitative real-time RT-PCR analysis for the matrix gene


of influenza A virus and the H3 gene of canine influenza A (H3N8).










Primer
Target
Sequence
Application





Ca-H3-F387
H3 (nt 387-406)
5′-tatgcatcgctccgatccat-3′
Forward primer for H3




(SEQ ID NO: 79)






Ca-H3-R487
H3 (nt 487-467)
5′-gctccacttcttccgttttga-3′
Reverse primer for H3




(SEQ ID NO: 80)






Ca-H3-P430
H3 (nt 430-459)
FAM-aattcacagcagagggattcacatggacag-BHQ1
TaqMan ® probe




(SEQ ID NO: 81)






FluA-M-F151
M (nt 151-174)
5′-catggartggctaaagacaagacc-3′a
Forward primer for M




(SEQ ID NO: 82)






FluA-M-R276
M (nt 276-253)
5′-agggcattttggacaaakcgtcta-3′
Reverse primer for M




(SEQ ID NO: 83)






FluA-M-P218
M (nt 218-235)
FAM-acgcTcaccgTgcccAgt-BHQ1b
TaqMan ® probe




(SEQ ID NO: 84)






aUnderlined letter r represents nucleotide a or g and underlined letter k represents nucleotide g or t.




bUppercase letters represent locked nucleic acid residues.














TABLE 8







Quantitative real-time RT-PCR and viral isolation performed on


lung tissues from dogs that died from pneumonia during respiratory


disease outbreaks in a shelter and veterinary clinic in Florida.













Duration of
Real-time





clinical
RT-PCR
Virus












Dog ID
Location
disease
M (Ct)
HA (Ct)
Isolation













A/canine/FL/242/03 positive control
28.15
27.36













1079
Shelter (NE FL)
 2 days
29.81
28.84
none


1078
Shelter (NE FL)
 3 days
30.37
29.71
MDCK







3rd passage


 318
Shelter (NE FL)
 9 days
33.89
32.97
none


 320
Shelter (NE FL)
10 days
39.44
37.09
none


 319
Shelter (NE FL)
 6 days
33.87
32.23
none


1080
Shelter (NE FL)
 6 days
38.87
38.23
none


 374
Veterinary clinic
 3 days
24.05
22.65
Egg



(SE FL)



2nd passage
















TABLE 9







Amino acid comparison of the mature HA for canine influenza viruses and contemporary equine influenza viruses.









Amino Acid


























7
15
54
78
79
83
92
107
118
159
218
222
261
328
479
483
492
541





A/equine/KY/5/02
G
I
N
V
F
N
S
S
L
N
G
W
K
I
G
N
R
K


A/equine/MA/213/03
.
.
.
A
.
.
.
.
.
S
.
.
.
.
.
.
.
.


A/equine/OH/1/03
D
.
K
A
.
.
.
.
.
S
.
.
.
.
.
.
.
.


A/canine/FL/242/03
.
M
K
A
.
S
.
.
.
S
.
L
.
T
.
T
.
.


A/canine/FL/43/04
.
M
K
A
.
S
N
.
.
S
.
L
.
T
.
T
.
R


A/canine/TX/1/04
.
M
K
A
.
S
.
.
V
S
.
L
N
T
E
T
.
.


A/canine/Iowa/05
.
M
K
A
L
S
.
.
V
S
E
L
N
T
E
T
.
.


A/canineMiami/05
.
M
K
A
L
S
.
.
V
S
E
L
N
T
E
T
K
.


A/canine/Jacksonville/05
.
M
K
A
L
S
.
P
V
S
E
L
N
T
E
T
K
.
















TABLE 10







Antibody titers in equine, canine, and ferret


serum to older and contemporary equine influenza viruses


and canine influenza viruses.









Serum antibody titersa










Antigens
Equine
Canine
Ferretb













equine/Miami/63
40
<10
16


equine/Ky/86
40
40
32


equine/KY/92
40
<10
32


equine/NY/99
320
40
128


equine/KY/05/02
320
160
256


equine/MA/213/03
640
160
512


equine/OH/01/03
640
160
512


canine/FL/03
160
160
512


canine/FL/04
160
80
512


canine/Tx/04
160
160
512


canine/Miami/05
160
80
256


canine/Jax/05
40
40
128






aAntibody titers were determined in a hemagglutination inhibition assay performed with serial dilutions of equine, canine, or ferret serum and the viruses listed in the antigen column.




bSerum from ferrets immunized with canine/FL/04 virus.







Materials and Examples Methods for Examples 12-15

Canine Influenza Virus inoculum.


The virus inoculum was prepared by inoculation of Madin-Darby canine kidney (MDCK) epithelial cells with a stock of A/canine/FL/43/04 (H3N8) representing passage 3 of The original isolate previously described (Crawford et al., 2005). The inoculated MDCK cells in Dulbecco's Minimal Essential Media (DMEM) supplemented with 1 μg/mL TPCK-treated trypsin (Sigma-Aldrich Corp., St. Louis, MO) and antibiotics (gentamycin and ciprofloxacin) were grown in 250 cm2 flasks at 37° C. in a humidified atmosphere containing 5% CO2. The cultures were observed daily for morphologic changes and harvested at 5 days post inoculation. The harvested cultures were clarified by centrifugation and the supernatants were stored at −80° C. pending inoculation of dogs. An aliquot of supernatant was used for determination of virus titer by the Reed and Muench method. The titer was 107 median tissue culture infectious doses (TCID50) of A/canine/Florida/43/2004 (canine/FL/04) per mL.


Experimental Inoculation.


Eight 4-month old colony bred mongrel dogs (Marshall BioResources, North Rose, NY) (4 males and 4 females) were used for the experimental inoculation study approved by the University of Florida Institutional Animal Care and Use Committee. The dogs' body weights ranged from 13 to 17 kg. The dogs were healthy based on physical examinations, baseline blood tests, and recording of body temperatures for 2 weeks prior to inoculation. All dogs were free from prior exposure to canine influenza virus based on serology tests performed on paired serum samples collected at the time of arrival into the facility and 2 weeks later. The dogs were anesthetized by intravenous injection of propofol (Diprivan®, Zeneca Pharmaceuticals, 0.4 mg/kg body weight to effect) for intubation with endotracheal tubes. Six dogs (3 males and 3 females) were each inoculated with 107 TCID50 of canine/FL/04 virus in 5 mL of sterile saline administered into the distal trachea through a small diameter rubber catheter inserted into the endotracheal tube. Two dogs (1 male and 1 female) were sham-inoculated with an equal volume of sterile saline. The sham-inoculated control dogs were housed in a different room from the virus-inoculated dogs and cared for by different personnel. Physical examinations and rectal temperature recordings were performed twice daily for 6 days post inoculation (p.i.).


Pharyngeal and Rectal Swab Collection.


To monitor for virus shedding, oropharyngeal specimens were collected twice daily from each dog on days 0 to 6 p.i. using polyester swabs (Fisher Scientific International Inc., Pittsburgh, PA). The swabs were placed in 1 mL of sterile phosphate-buffered saline (PBS) containing 0.5% bovine serum albumin (BSA). Rectal swabs were collected from each dog daily from days 0 to 6. Swab extracts were prepared by clarification of the swab transport media by centrifugation. An aliquot of swab extract was tested immediately for influenza A virus nucleoprotein using the Directigen™ commercial immunoassay kit (BD, Franklin Lakes, NJ) according to the manufacturer's instructions. The remaining extract was stored at −80° C. pending other virological assays.


Postmortem Examinations.


On day 1 p.i., one sham-inoculated dog and one virus-inoculated dog were euthanatized by intravenous inoculation of Beuthanasia-D® solution (1 mL/5 kg body weight; Schering-Plough Animal Health Corp). One virus-inoculated dog was similarly euthanatized each day from days 2 to 5 p.i. On day 6 p.i., the remaining sham-inoculated and virus-inoculated dog were euthanatized. Complete postmortem examinations were performed by one of the investigators (WLC). Tissues were fixed in 10% neutral buffered formalin, embedded in paraffin, and 5-μm sections were either stained with hematoxylin and eosin for histopathologic diagnosis or processed for immunohistochemistry as described below. Unfixed lung tissues were submitted to the Diagnostic Clinical Microbiology/Parasitology/Serology Service at the University of Florida College of Veterinary Medicine for bacterial isolation and identification. The samples were cultured on nonselective media as well as media selective for Bordetella species (Regan-Lowe; Remel, Lenexa, KS) and Mycoplasma species (Remel). All cultures were held for 21 days before reporting no growth. Unfixed tissues were also stored at −80° C. pending virological analyses.


Immunohistochemistry.


Deparaffinized and rehydrated 5 μm trachea and lung tissue sections were mounted on Bond-Rite™ slides (Richard-Allan Scientific, Kalamazoo, MI) and subsequently treated with proteinase K (DAKOCytomation Inc., Carpenteria, CA) followed by peroxidase blocking reagent (DAKO® EnVision™ Peroxidase Kit, DAKO Corp., Carpenteria, CA). The sections were incubated with a 1:500 dilution of monoclonal antibody to influenza A H3 (Chemicon International, Inc., Ternecula, CA) for 2 hours at room temperature. Controls included incubation of the same sections with mouse IgG (1 mg/mL, Serotec, Inc. Raleigh, NC), and incubation of the monoclonal antibody with normal canine lung sections. Following treatment with the primary antibody, the sections were incubated with secondary immunoperoxidase and peroxidase substrate reagents (Dako® EnVision™ Peroxidase Kit, Dako Corp.) according to the manufacturer's instructions. The sections were counterstained with hematoxylin, treated with Clarifier #2 and Bluing Reagent (Richard-Allan Scientific, Kalamazoo, MI), dehydrated, and coverslips applied with Permount (ProSciTech, Queensland, Australia).


RNA Extraction from Swabs and Tissues.


Lung and tracheal tissues from each dog were thawed and homogenized in minimum essential medium (MEM) supplemented with 0.5% bovine serum albumin (BSA) and antibiotics (gentamycin and ciprofloxacin) using a disposable tissue grinder (Kendall, Lifeline Medical Inc., Danbury, CT). Total RNA was extracted from the tissue homogenates as well as orpharyngeal and rectal swab extracts using a commercial kit (RNeasy® Mini Kit, QIAGEN Inc., Valencia, CA) according to manufacturer's instructions and eluted in a final volume of 60 μL of buffer.


Real-Time RT-PCR.


A single-step quantitative real-time RT-PCR was performed on the total RNA using the QuantiTect® Probe RT-PCR Kit containing ROX as a passive reference dye (QIAGEN Inc., Valencia, CA) and a primer-probe set that targeted a highly conserved region of the matrix (M) gene of type A influenza virus (Payungporn S. et al., 2006a; Payungporn S. et al., 2006b). For each real-time RT-PCR reaction, 5 μL of extracted total RNA were added to a reaction mixture containing 12.5 μL of 2× QuantiTech® Probe RT-PCR Master Mix, 0.25 μL of QuantiTech® RT Mix, forward and reverse primers (0.4 μM final concentration for each), probe (0.1 μM final concentration) and RNase-free water in a final volume of 25 μL. The TaqMan® GAPDH Control Reagents (Applied Biosystems, Foster City, CA) were used according to manufacturer's instructions for detection of GAPDH as an endogenous internal control for the presence of RNA extracted from the swab and tissue samples and as a normalization control.


Quantitative one-step real-time RT-PCR was performed on the reaction mixtures in a Mx3000P® QPCR System (Stratagene, La Jolla, CA). Cycling conditions included a reverse transcription step at 50° C. for 30 minutes, an initial denaturation step at 95° C. for 15 minutes to activate the HotStarTaq® DNA polymerase, and amplification for 40 cycles. Each amplification cycle included denaturation at 94° C. for 15 seconds followed by annealing/extension at 60° C. for 1 minute. The FAM (emission wavelength 518 nm) and VIC (emission wavelength 554 nm) fluorescent signals were recorded at the end of each cycle. The threshold cycle (Ct) was determined by setting the threshold fluorescence (dR) at 1000 in each individual experiment. The Mx3000P® version 2.0 software program (Stratagene, La. Jolla, CA) was used for data acquisition and analysis. The positive control consisted of amplification of RNA extracted from A/canine/FL/242/03 (H3N8) virus. The results were normalized by dividing the M Ct value by the corresponding GAPDH Ct value for each sample.


Virus Re-Isolation from Tissues.


Frozen lung and trachea tissues from virus-inoculated dogs were thawed and homogenized in 10 volumes of DMEM supplemented with 0.5% BSA and antibiotics. Solid debris was removed by centrifugation and supernatants were inoculated onto MDCK cells cultured in DMEM supplemented with 1 μg/mL TPCK-treated trypsin (Sigma-Aldrich Corp., St. Louis, MO) and antibiotics as described above. Cells were grown in 25 cm2 flasks at 37° C. in a humidified atmosphere containing 5% CO2. The cultures were observed daily for morphologic changes and harvested at 5 days post inoculation. The harvested cultures were clarified by centrifugation and the supernatants inoculated onto fresh MDCK cells as described for the initial inoculation; two additional passages were performed for samples that did not show evidence of influenza virus by hemagglutination or RT-PCR. Hemagglutination activity in the clarified supernatants was determined using 0.5% turkey red blood cells as previously described (Crawford et al., 2005). RT-PCR was performed as described below.


RT-PCR, Nucleotide Sequencing, and Phylogenetic Analyses.


Viral RNA was extracted from MDCK supernatant using the QIAamp® Viral RNA Mini Kit (QIAGEN Inc., Valencia, CA) according to manufacturer's instructions. The viral RNA was reverse transcribed to cDNA using the QIAGEN® OneStep RT-PCR Kit (QIAGEN Inc., Valencia, CA) according to manufacturer's instructions. PCR amplification of the coding region of the 8 influenza viral genes in the cDNA was performed as previously described (Crawford et al., 2005), using universal gene-specific primer sets (primer sequences available on request). The resulting DNA amplicons were used as templates for automated sequencing in the ABI PRISM® 3100 automated DNA sequencer using cycle sequencing dye terminator chemistry (Applied Biosystems, Foster City, CA). Nucleotide sequences were analyzed using the Lasergene 6 Package® (DNASTAR, Inc., Madison, WI). The nucleotide sequences for viruses recovered from infected dogs were compared to the sequences of the virus in the inoculum to determine if any changes had occurred during replication in the respiratory tract.


Example 12-Clinical Disease

All 6 virus-inoculated dogs developed a transient fever (rectal temperature ≥39° C.) for the first 2 days p.i., but none exhibited respiratory signs such as cough or nasal discharge over the 6-day observation period. The sham-inoculated dogs remained clinically healthy.


Example 13-Virus Shedding

Influenza A nucleoprotein was detected in the oropharyngeal swab collected from one of the virus-inoculated dogs at 24 hours p.i. The oropharyngeal swabs collected from one dog at 72, 84, and 120 hours p.i., and another dog at 108, 120, and 132 hours p.i., were positive for virus by quantitative real-time RT-PCR (Table 11). The absolute number of influenza M gene copies per μL of swab extract increased with time from 3 to 6 days p.i. No virus was detected in the rectal swabs.


Example 14-Postmortem Examinations

In contrast to the previous experimental infection using specific pathogen-free Beagles (Crawford et al., 2005), the virus-inoculated mongrel dogs had pneumonia as evidenced by gross and histological analyses of the lungs from days 1 to 6 p.i. In addition to pneumonia, the dogs had rhinitis, tracheitis, bronchitis, and bronchiolitis similar to that described in naturally infected dogs (Crawford et al., 2005). There was epithelial necrosis and erosion of the lining of the airways and bronchial glands with neutrophil and macrophage infiltration of the submucosal tissues (FIG. 5, upper panels). Immunohistochemistry detected viral H3 antigen in the epithelial cells of bronchi, bronchioles, and bronchial glands (FIG. 5, lower panels). No bacterial superinfection was present. The respiratory tissues from the 2 sham-inoculated dogs were normal.


Example 15-Virus Replication in Trachea and Lungs

The trachea and lungs were positive for virus by quantitative real-time RT-PCR in all dogs from 1 to 6 days p.i. (Table 12). The absolute number of influenza M gene copies per μL of trachea homogenate increased from 1 to 5 days p.i., then decreased on day 6. The absolute number of M gene copies per μL of lung homogenate decreased from 1 to 6 days p.i. In general, the trachea contained ≥ one log10 more virus than the lung on each of the 6 days p.i.









TABLE 11







Detection of virus shedding in the oropharynx of


inoculated with canine influenza virus mongrel dogs


by quantitative real-time RT-PCR.













Time p.i.

Matrix gene



Dog ID
(hours)a
M/GAPDH ratiob
(copies/uL)c







860
 72
1.20
1.57E+02




 84
1.30
8.25E+02




120
1.23
1.47E+03



894
108
1.17
1.17E+02




120
1.41
1.37E+02




132
1.27
3.74E+02








aTime that oropharyngeal swabs were collected from the dogs following inoculation with A/canine/FL/43/04 (H3N8) virus.





bNormalization ratios were calculated by dividing the M (Ct) by the GAPDH (Ct) for each swab extract.





cThe absolute number of matrix gene copies per uL of swab extract.














TABLE 12







Detection of virus replication in the trachea and lung of


mongrel dogs inoculated with canine influenza virus by


quantitative real-time RT-PCR.












M/GAPDH
Matrix gene



Time p.i.
ratiob
(copies/uL)c












Dog ID
(hours)a
Lung
Trachea
Lung
Trachea















797
24
1.20
1.43
8.22E+05
3.11E+04


801
48
1.33
0.99
1.15E+05
6.52E+06


789
72
1.44
1.12
2.39E+04
1.56E+05


819
96
1.40
1.27
3.19E+04
1.43E+05


860
120
1.59
1.04
3.48E+03
1.17E+06


894
144
1.70
1.15
4.78E+02
1.50E+03






aTime that tissues were collected from the dogs following inoculation with A/canine/FL/43/04 (H3N8) virus.




bNormalization ratios were calculated by dividing the M (Ct) by the GAPDH (Ct) for each tissue homogenate.




cThe absolute number of matrix gene copies per uL of tissue homogenate.







Materials and Examples Methods for Example 16

Virus Strains


Canine influenza virus strains as well as those of avian, equine and human origin (listed in Table 15) were propagated in embryonated eggs or MDCK cells and their infectivity was titrated by endpoint dilution in chicken embryos, or plaque assay. Rapid virus quantification was performed by hemagglutination assay using turkey red blood cell erythrocytes.


Diagnostic Specimens


A Total of 60 canine's lung tissues collected from suspect cases of viral respiratory disease during the year of 2005 were tested for the presence of canine influenza virus.


RNA Extraction from Canine Tissue Samples


Blocks of lung tissue weighing between 20 and 30 mg were homogenized in a disposable tissue grinder (Kendal). Total RNA was extracted using a commercial kit (RNeasy Mini Kit, Qiagen, Valencia, CA) and eluted in a final volume of 60 μL, following the manufacturer's recommendations.


Primers and Probes Design


Multiple sequence alignments of the H3 and M genes from various subtypes and from diverse species were performed using the CLUSTAL X program (Version 1.8). Matrix (M) primers and probe were selected from the conserved regions of over the known sequences corresponding to different subtypes of influenza A virus, whereas the H3 hemagglutinin gene-specific primers and probe set were selected to specifically match equine and canine influenza A virus genes and mismatch the homologous avian and human genes (Table 13). Primer design software (OLIGOS Version 9.1) and the web based analysis tools provided by EXIQON (http://lnatools.com) was used for Tm calculation and prediction of secondary structure as well as self hybridization. A conserved region of an 18S rRNA gene was used as endogenous internal control for the presence of RNA extracted from canine tissue sample. The Pre-Developed TaqMan® Assay Reagents for Eukaryotic 18S rRNA (VIC/TAMRA) (Applied Biosystems) was used for the real-time detection of 18S rRNA in tissue samples.


Real-time RT-PCR Condition


A single-step real-time RT-PCR was performed by using the Quantitect Probe RT-PCR time RT-PCR reaction, 5 μL of RNA sample were used as a template to combine with a reaction mixture containing 10 μL of 2× QuantiTech Probe RT-PCR Master Mix, 0.2 μL of QuantiTech RT Mix, primers (0.4 μM final conc. for H3 gene or 0.6 μM final conc. for M gene), probe (0.1 μM final conc. for H3 gene or 0.2 μM final conc. for M gene) and RNase-free water in a final volume of 20 μL. One-step real-time RT-PCR was performed in the Mx3005P Real-Time QPCR System (Stratagene). Cycling conditions included a reverse transcription step at 50° C. for 30 minutes. After an initial denaturation step at 95° C. for 15 minutes in order to activate the HotStarTaq DNA polymerase, amplification was performed during 40 cycles including denaturation (94° C. for 15 seconds) and annealing/extension (60° C. for 30 seconds). The FAM (emission wavelength 516 nm for H3 and M detection) and VIC (emission wavelength 555 nm for 18S rRNA detection) fluorescent signals were obtained once per cycle at the end of the extension step. Data acquisition and analysis of the real-time PCR assay were performed using the Mx3005P software version 2.02 (Stratagene).


Specificity of H3 Primers/Probe Set for Canine Influenza (H3N8) and Universality of M Primers/Probe Set for Type A Influenza Virus


In order to test the specificity of each primers/probe set, RNA extracted from several known subtypes of influenza A viruses were used as a template in the real-time RT-PCR assay (Table 15).


RNA Standard for Determination of the Real-Timer RT-PCR Performance


The genes of canine influenza A virus (A/canine/Florida/242/2003(H3N8)) were used to generate the PCR amplicons for H3 (nt 1-487) and M (nt 1-276) by using primers linked with T7 promoter (Table 13). Then the purified PCR amplicons of H3 and M genes were used as templates for in vitro transcription by using Riboprobe in vitro Transcription System-T7 (Promega) following the manufacturer's recommendations. The concentration of the transcribed RNAs was calculated by measuring absorbance at 260 nm. The RNAs were then serially diluted 10-fold, ranging from 108 to 10 copies/μL to perform sensitivity tests. Moreover, a standard curve was generated by plotting the log of initial RNA template concentrations (copies/μL) against the threshold cycle (Ct) obtained from each dilution in order to determine the overall performance of real-time RT-PCR.


Comparative Sensitivity Tests Between Real-Time RT-PCR and Directigen Flu A Test Kit


Stock viruses of two viral strains including A/Wyoming/3/2003 (H3N2) at 106.67 EID50/mL (HA=64) and A/canine/Florida/242/2003(H3N8) at 107.17 EID50/mL (HA=16) were used for the detection threshold assay. Logarithmic dilution of specimens in phosphate-buffered saline (PBS) (125 μL) were used in a rapid influenza A antigen detection kit, Directigen Flu A, (Becton, Dickinson and Company) following the manufacturer's instructions. Each Directigen Flu A test device has an H1N1 influenza antigen spot in the center of the membrane which develops as a purple dot and indicates the integrity of the test, which is based on a monoclonal antibody to the nucleoprotein (NP). The development of a purple triangle surrounding the dot is indicative of the presence of influenza NP in the tested specimen. The intensity of the purple signal from the triangle was scored as + (outline of triangle), ++ (lightly colored triangle), +++ (dark-purple triangle) and ++++ (very dark-purple triangle). Viral RNA was extracted 125 μL aliquots of each virus dilution by using QIAamp Viral RNA Mini Kit (Qiagen, Valencia, CA) and eluting in a final volume of 50 μL. A volume of 5 uL of the extracted viral RNAs were tested by real-time RT-PCR for comparative sensitivity test with Directigen Flu A kit.


Example 16

The real-time RT-PCR assay for canine influenza relies on information from three molecular probes which target 18S rRNA from host cell was well as M and H3 from the influenza A virus genome (Table 14). Amplification of the host gene is a reporter of specimen quality and integrity. Clinical, necropsy or laboratory samples containing canine influenza (H3N8) virus are expected to yield amplification signal with the three probes. Specimens yielding amplification signal with M and 18S rRNA probes but negative for H3 would be indicative of an influenza virus subtype H3 from human, swine or avian origin or from non-H3 subtypes. These rare cases could be resolved by RT-PCR using HA universal primers to generate amplicon cDNA that can be analyzed by sequencing. Properly collected and handled specimens lacking influenza A virus yield 18S rRNA amplification signal only. Situations in which only the 18S rRNA probe and the H3 probes yield amplification signal are indicative of faulty technique, unless proven otherwise; either a false negative with M probes or false positive for H3 need to be demonstrated. Finally, specimens failing to yield amplification signals with the three probes are indicative of defective sample collection, degradation, faulty RNA extraction or the presence of inhibitors the polymerases used in PCR.


In order to test the specificity of the H3 primers/probe set for canine influenza A virus (H3N8) and the universality of M primers/probe set for type A influenza, several subtypes of influenza A viruses were tested by real-time RT-PCR. The results show that H3 primers/probe set yielded a positive amplification signal only with canine influenza (H3N8). No significant false positive or non-specific amplification signals were observed in other subtypes or human H3 strains. The M primers/probe set yielded positive amplification signal with all of the strains tested (Table 15). These results indicated that H3 primers/probe specifically detects canine influenza A virus (H3N8) whereas M primers/probe detect multiple subtypes of type A influenza viruses.


The performance of real-time RT-PCR assays was evaluated by endpoint dilution of M and H3 in vitro transcribed RNAs. As expected, the threshold cycle (Ct) increased in direct correlation with the dilution of the RNA standards. The fluorescent signals can be detected at RNA standard dilutions of M and H3 as low as 103 and 102 copies/μL, respectively (FIGS. 6A and 6B). The standard curves of M and H3 genes were constructed by plotting the log of starting RNA concentrations against the threshold cycle (Ct) obtained from each dilution (FIGS. 6C and 6D). The slope of the standard curve is used to determine the PCR reaction efficiency, which is theoretically exponential; 100% amplification efficiency would imply doubling of amplicon concentration each cycle. The standard curves with a slope between approximately −3.1 and −3.6 are typically acceptable for most applications requiring accurate quantification (90-110% reaction efficiency). An Rsq value is the fit of all data to the standard curve plot. If all the data lie perfectly on the line, the Rsq will be 1.00. As the data fall further from the line, the Rsq decreases. An Rsq value ≥0.985 is acceptable for most assays. The M standard curve revealed a slope of −3.576 (efficiency=90.4%) and Rsq=1.00 whereas H3 standard curve yielded a slope of −3.423 (efficiency=95.9%) and Rsq=0.999. These values indicate satisfactory amplification efficiency and overall performance of the real-time RT-PCR assays. We attribute the lower efficiency and sensitivity of M primers/probe set as compared to H3 primers/probe set to the N-fold degeneracy of M primer sequences required to ensure broad coverage of M gene sequences variability across viruses of multiple subtypes, hosts and lineages.


The sensitivity of real-time RT-PCR assay was also compared with the commercial rapid antigen detection assay (Directigen Flu A). Logarithmic dilutions of A/Wyoming/3/2003 (H3N2) and A/canine/Florida/242/2003(H3N8) were analyzed with Directigen Flu A and by real-time RT-PCR. The results of Directigen Flu A showed that the sensitivities against both viral strains are approximately 100-fold dilution from the stock viruses used in these experiments (FIG. 7). The signals (purple color) generated by the canine virus (A/canine/Florida/242/2003: 106.x(PFU/ml) samples were much weaker than those found in human virus (A/Wyoming/3/2003: 107.x PFU/ml), in agreement with the lower virus concentration in these samples. Alternatively, lower signal for canine influenza could be attributed to the molecular specificity of monoclonal antibodies against the NP; i.e. poor conservation of the amino acids within the NP epitope of canine influenza A viruses.


Real-time RT-PCR of the M gene yielded Ct values above threshold with virus 10 and 30 PFU equivalents per reaction of A/canine/Florida/242/2003 and A/Wyoming/3/2003, respectively (Table 16). The differences between the sensitivity value of 2 viral strains because the differences in the original viral titers. The H3 gene detection comparison between canine and human influenza viruses was not performed because the H3 primers/probe in our realtime RT-PCR assay amplifies exclusively canine influenza A virus. RT-PCR was 105 times more sensitive than the rapid antigen detection kit.


To evaluate the performance of the RT-PCR test in necropsy specimens from dogs with acute respiratory disease, sixty canine lung tissue samples submitted during the year of 2005 were tested for the presence of canine influenza A virus by real-time RT-PCR. A total of 12 out of 60 samples (20%) were positive with both M and H3 genes whereas the remaining 48 samples yielded negative result for both M and H3 gene. Virus isolation attempts were conducted by egg and MDCK cell inoculation, to evaluate the specificity of the realtime assay; 2 out 12 samples that were positive for canine influenza by RT-PCR yielded canine influenza virus (data not shown, manuscript in preparation). Although all of the tissues were collected from dogs with a history of severe respiratory disease, most of the samples yielded no canine influenza virus by either realtime PCR or conventional isolation, suggesting a high incidence of other respiratory pathogens such as Bordetella bronchiseptica, canine distemper or parainfluenza virus. The single step real-time RT-PCR assay herein provides a rapid, sensitive and cost-effective approach for canine influenza A virus (H3N8) detection. Rapid laboratory diagnosis of canine influenza A virus (H3N8) infections in the early stage of the disease can yield information relevant to clinical patient and facility management.









TABLE 13







Primers and probes used for real-time RT-PCR detection and in vitro transcription











Oligo name
Type
Target
Sequence*
Application





Ca-H3-
Forward
H3 (nt 387-406)
5′-tatgcatcgctccgatccat-3′
Real-time


F387
primer

(SEQ ID NO: 79)
PCR


Ca-H3-
Reverse
H3 (nt 487-467)
5′-gctccacttcttccgttttga-3′



R487
primer

(SEQ ID NO: 80)



Ca-H3-
TaqMan
H3 (nt 430-459)
FAM-aattcacagcagagggattcacatggacag-BHQ1



P430
probe

(SEQ ID NO: 81)






FluA-M-
Forward
M (nt 151-174)
5′-catggartggctaaagacaagacc-3′
Real-time


F151
primer

(SEQ ID NO: 82)
PCR


FluA-M-
Reverse
M (nt 276-253)
5′-agggcattttggacaaakcgtcta-3′



R276
primer

(SEQ ID NO: 83)



FluA-M-
LNA
M (nt 218-235)
FAM-acgcTcaccgTgcccAgt-BHQ1



P218
TaqMan

(SEQ ID NO: 84)




probe








H3-F1
Forward
H3 (nt 1-14)
5′-tattcgtctcagggagcaaaagcagggg-3′
In vitro



primer

(SEQ ID NO: 85)
transcription


T7/H3-
Reverse
T7/H3
5′-tgtaatacgactcactatagggctccacttcttccgttttga-3′



R490
primer
(nt 487-467)
(SEQ ID NO: 86)






M-F1
Forward
M (nt 1-15)
5′-gatcgctcttcagggagcaaaagcaggtag-3′
In vitro



primer

(SEQ ID NO: 87)
transcription


T7/M-R276
Reverse
M (nt 276-253)
5′-tgtaatacgactcactatagggcattttggacaaagcgtc-3′




primer

(SEQ ID NO: 88)





*Note:


Uppercases = LNA (Locked Nucleic Acid) residues,


r = a or g,


k = g or t,


underline = T7 promoter sequence













TABLE 14







Interpretation of the real-time RT-PCR assay









Results










Interpretation
M
H3
18S rRNA





Positive for canine influenza A virus (H3N8)
+
+
+


Positive for influenza A virus
+

+


(unknown subtype)





Negative for influenza A virus


+


Error in RNA extraction or presence of





PCR inhibitor
















TABLE 15







Specificity test of canine H3 primers/probe set and universality test of


M primers/probe set with several subtypes of influenza A viruses













Real-time RT-PCR





detection














H3 gene
M gene


Subtypes
Strain Name
Host
(Ct)
(Ct)














H1 
A/Ohio/1983
Human
No Ct
15.40



A/WSN/1933
Human
No Ct
20.09


H3 
A/Wyoming/3/2003
Human
No Ct
28.85



A/Victoria/3/1975
Human
No Ct
16.62



A/canine/FL/242/2003
Canine
28.43
29.25


H4 
Turkey/MN/1066/1980
Avian
No Ct
17.49



Clinical sample*
Avian
No Ct
20.87


H5 
AChicken/Thailand/CUK2/2004
Avian
No Ct
20.13



A/Pheasant/NJ/1335/1998
Avian
No Ct
16.64


H6 
Clinical sample*
Avian
No Ct
19.52


H10
Clinical sample*
Avian
No Ct
25.64



Clinical sample*
Avian
No Ct
19.59


H11
Clinical sample*
Avian
No Ct
15.72



Clinical sample*
Avian
No Ct
24.55





*Note


that subtypes of clinical samples were confirmed by nucleotide sequencing.













TABLE 16







Comparative sensitivity tests for influenza A virus detection


between real-time RT-PCR and Directigen Flu A










Directigen Flu A
Real-time RT-PCR of M (Ct)











Virus
A/canine/
A/Wyoming/
A/canine/
A/Wyoming/


dilutions
242/03
3/03
242/03
3/2003





10−1
++
++++
22.42
19.48


10−2
+
+++
25.85
22.66


10−3


29.27
25.76


10−4
Not done
Not done
32.66
28.66


10−5
Not done
Not done
35.48
33.14


10−6
Not done
Not done
37.51
35.06


10−7
Not done
Not done
39.09
36.44


10−8
Not done
Not done
No Ct
38.93



















TABLE 17







Class of Amino Acid
Examples of Amino Acids









Nonpolar
Ala, Val, Leu, Ile, Pro, Met, Phe, Trp



Uncharged Polar
Gly, Ser, Thr, Cys, Tyr, Asn, Gln



Acidic
Asp, Glu



Basic
Lys, Arg, His




















TABLE 18







Letter Symbol
Amino Acid









A
Alanine



B
Asparagine or




aspartic acid



C
Cysteine



D
Aspartic Acid



E
Glutamic Acid



F
Phenylalanine



G
Glycine



H
Histidine



I
Isoleucine



K
Lysine



L
Leucine



M
Methionine



N
Asparagine



P
Proline



Q
Glutamine



R
Arginine



S
Serine



T
Threonine



V
Valine



W
Tryptophan



Y
Tyrosine



Z
Glutamine or




glutamic acid

















TABLE 19







Amino acid differences between PB2 proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04
















5
K
K
E



12
S
L
L



37
G
G
E



175
R
R
I



374
L
I
I



375
R
R
K



447
Q
Q
H

















TABLE 20







Amino acid differences between PB1 proteins


of H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04







114
V
I
I



154
D
G
G



221
A
T
T



317
M
I
I



459
I
I
V



682
I
I
V

















TABLE 21







Amino acid differences between PA proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04
















27
D
N
N



62
I
V
V



213
R
K
K



337
A
T
T



343
A
E
E



345
L
I
I



353
K
R
R



400
T
T
A



450
V
I
I



460
M
M
I



673
R
R
K



675
N
D
D







*Based on available genes of viruses isolated between 1963 and 1998.













TABLE 22







Amino acid differences between NP proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04
















16
G
D
D



157
A
T
T



214
R
R
K



285
V
V
I



286
A
T
T



359
A
T
T



375
D
D
N



384
R
K
K



452
R
K
K

















TABLE 23







Amino acid differences between NA proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04
















9
A/T
T
A



12
S
F
F



20
L
I
I



40
G
R
R



42
G
D
D



46
N
K
K



52
E
E
K



61
R
K
K



69
N
S
S



72
E
K
K



201
V
I
I



261
I
V
V



301
I
I
V



396
N
D
D



397
L
P
P

















TABLE 24







Amino acid differences between M1 proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04







M1 161
S
S
A



M1 208
K/Q
R
R







*Based on available genes of viruses isolated between 1963 and 1998.













TABLE 25







Amino acid differences between NS1 proteins of


H3N8 equine and canine influenza viruses












Position
Equine Consensus *
Canine/FL/03
Canine/FL/04
















44
K
R
R



59
R
H
H



71
E
K
K



86
A
T
T



88
R
R
L



140
R
G
G



216
P
S
S







*Based on available genes of viruses isolated between 1963 and 1998.






Example 17—Canine Influenza Challenge Model Development

The canine influenza (canine flu) virus, which was isolated from flu outbreaks in Florida, was observed to be a H3N8 type influenza virus, and closely related to equine flu virus strain, A/equine/Ohio/03 (Crawford et al., SCIENCE Vol. 309, September 2005, incorporated by reference in its entirety into this patent). The potential of using the equine flu virus strain A/equine/Ohio/03 to induce influenza-like disease in dogs was investigated in this study.


Procedure:


Ten 13-week-old beagles of mixed sex were obtained from a commercial supplier, and housed in individual cages in a BSL-2 facility. The dogs were randomly assigned to two groups of 5 dogs each. As shown in Table 26, one group was subjected to a intratracheal challenge, and the other group was subjected to an oronasal challenge. The dogs were challenged at 14 weeks-of-age.









TABLE 26







Experimental Design











Group
Number of Dogs
Challenge Route







1
5
Intratracheal



2
5
Oronasal










A cell culture grown equine flu virus A/equine/Ohio/03 was used as the challenge virus. For intratracheal challenge, the challenge virus was administered via a delivery tube, which consisted of a cuffed tracheal tube (Size 4.0/4.5, Sheridan, USA) and feeding tube (size 5 Fr, 1.7 mm, /16 inches in length, Kendall, USA) in 0.5 to 1.0 ml volume. For oronasal challenge, the challenge virus (107 to 108 TCID50 per dog) was administered as a mist using a nebulizer (DeVilbiss ULTRA-NEB 99 ultrasonic nebulizer, Sunrise Medical, USA) in a 2 to 3 ml volume.


The dogs were observed for flu related clinical signs for 14 days post-challenge. Serum samples were collected from each dog on day zero (before challenge), and days 7 and 14 post-challenge for determining the HI titer using a H3N8 equine influenza virus with a standard protocol (SAM 124, CVB, USDA, Ames, IA). All dogs were humanely euthanized and lung tissues were collected in 10% buffered formalin for histopathological evaluation.


Results:


The results of this experiment are summarized in Table 27. Influenza related clinical signs were observed in a few dogs after challenge. These signs included fever (>103° F.; >39.4° C.) and cough. Two of 5 dogs (i.e., 40%) had fevers (>103° F.; >39.4° C.) in Group 1, compared to 1 of 5 (i.e., 20%) dogs in Group 2. One dog from the oronasal challenge group had sneezing, and another had cough following the challenge. An HI titer range from 10 to 80, with a geometric mean titer (GMT) of 20, was observed for Group 1. A titer range of 40 to 160, with a GMT of 86, was observed for Group 2. One dog from each group had histopathological lesions compatible with or pathognomic for influenza.









TABLE 27







Canine flu challenge-clinical signs, virus isolation, histopathology results and serology results





















Serology (HI titer)

















Virus isolation
Microscopic

7-days
14-days

















Challenge
Clinical
Nasal/oral
Tracheal
Lung
lesion
Pre-
post
post


Dog* ID
method*
signs
swab
scraping
tissues
(histopathology)
challenge
challenge
challenge



















AAH
Intratracheal
none
negative
negative
negative
negative
10
10
20


ADB
Intratracheal
none
negative
negative
negative
negative
10
80
20


ADC
Intratracheal
Fever*
negative
negative
negative
negative
10
20
20


AEB
Intratracheal
Fever
negative
negative
negative
positive
10
40
20


AEE
Intratracheal
none
negative
negative
negative
inconclusive
10
20
10


AAE
Oronasal
none
negative
negative
negative
negative
10
80
80


AAG
Oronasal
none
negative
negative
negative
negative
10
40
80


ABY
Oronasal
Occasional
negative
negative
negative
positive
10
80
160




sneeze,











occasional











cough









ADY
Oronasal
Fever,
negative
negative
negative
negative
10
80
80




occasional











sneeze









ADZ
Oronasal
none
negative
negative
negative
negative
10
80
160





*The animals were challenged with an Equine flu isolate Ohio 03.


**Rectal temperature ≥103° F.; ≥39.4 ° C.






Example 18-Efficacy of an Equine Influenza Virus Vaccine for Dogs

The canine influenza (canine flu) virus isolated from flu outbreaks in Florida was observed to be a H3N8 type influenza virus, and was closely related to equine flu virus, A/equine/Ohio/03 based on the sequence similarity. The following study was conducted to determine the efficacy of an experimental inactivated equine influenza virus vaccine.


Procedure:


Nine 7-week-old beagles of mixed sex were obtained from a commercial supplier, and housed in individual cages in a BSL-2 facility. These dogs were randomly assigned to two groups, as summarized in Table 28:









TABLE 28







Experimental Design











Group
Number of Dogs
Treatment







1
5
Vaccine



2
4
Control











The first group consisted of 5 dogs, which were vaccinated with an inactivated, CARBIGEN™ (MVP Laboratories, Inc., Omaha, NE) adjuvanted, equine flu virus A/equine/Ohio/03 vaccine at 8 and 12 weeks-of-age via subcutaneous (SQ) route. The A/equine/Ohio/03 was inactivated by binary ethylenimine (“BEI”) using a standard method. Each dose of the vaccine contained 5% by mass CARBIGEN™, 4096 HA units of the inactivated virus, sufficient PBS to bring the total volume of the dose to 1 ml, and sufficient NaOH to adjust the pH to between 7.2 and 7.4. Serum samples were collected from all dogs on the day of first and second vaccination and day 7 and 14, post-first and -second vaccination, and at pre-challenge for determining the HI titer using a H3N8 equine influenza virus a standard protocol (SAM 124, CVB, USDA, Ames, IA). At 3 weeks post-second vaccination, the 5 vaccinated dogs and the second group (i.e., the control group) consisting of 4 age-matched dogs were challenged oronasally with a cell-culture-grown equine influenza virus A/equine/Ohio/03 (107.0 to 108.0 TCID50 per dog) in a 1-2 ml volume per dose. The challenge virus was administered to the dogs as a mist using a nebulizer (DeVilbiss ULTRA-NEB 99 ultrasonic nebulizer, Sunrise Medical, USA). The dogs were observed for flu-related clinical signs for 14 days post-challenge. Five dogs (3 vaccinates and 2 controls) 7 days post-challenge and 4 dogs (2 controls and 2 vaccinates) 14 days post-challenge were humanely euthanized for collection of lung tissues in 10% buffered formalin for histopathological evaluation.


Results:


The results of this experiment are summarized in Tables 29 and 30. All vaccinated dogs seroconverted following the vaccination. An HI titer range from 40 to 640, with the GMT of 129, was observed during the post-vaccination period with equine influenza virus A/equine/Ohio/03, and a HI titer of 160 to 320, with a geometric mean titer of 211, was observed with canine flu isolate, A/canine/Florida/242/03. Two of 6 vaccinates had a fever of >103° F. (>39.4° C.) for one day and no other clinical signs were observed in any of the dogs following challenge.


Conclusion:


All the vaccinated dogs responded to the inactivated, CARBIGEN™ adjuvanted equine influenza vaccine. The HI titer results with a canine influenza virus isolate suggest that the inactivated equine influenza vaccine did induce a detectable level of cross reactive antibody to canine influenza virus. Even though the challenge virus used in this did not induce any noticeable clinical disease in beagle dogs, based on the HI titer with a canine influenza virus isolate, it was concluded that inactivated equine vaccine could be used in dogs to induce cross reactive antibodies, which could potentially protect dogs against “canine flu” disease caused by H3N8 type canine influenza viruses.









TABLE 29







Serology - Pre- and post-vaccination and post-challenge HI titers











HI titers
















Post-1st
Post-2nd

















vaccination
vaccination
Post-challenge*
















Dog*
Group
Pre-vaccination
7-d
14-d
7-d
14-d
21-d
7-d
14-d



















AKT
Vaccinate**
<10
40
80
640
640
640
320
320


ALH
Vaccinate**
<10
40
80
320
160
160
80
***


ALU
Vaccinate**
<10
40
80
320
160
160
80
80


ANJ
Vaccinate**
<10
40
80
320
160
80
320
***


ANU
Vaccinate**
<10
40
80
320
160
80
160
***


AJW
Control
<10
<10
<10
<10
<10
<10
10
***


AKR
Control
<10
<10
<10
<10
<10
<10
10
***


ALZ
Control
<10
<10
<10
<10
<10
<10
20
20


ARC
Control
<10
<10
<10
<10
<10
<10
10
10





*The animals were challenged with an equine flu isolate Ohio 03


**CARBIGEN ™ adjuvanted inactivated equine flu virus Ohio 03 vaccine was used for vaccination


*** Euthanized 7-days post-challenge













TABLE 30







Canine flu challenge* - clinical signs,


virus isolation, histopathology results















Microscopic





Virus isolation
lesion













Dog
Treatment
Clinical
Nasal
Tracheal
Lung
(histo-


ID
group
signs
swab
scraping
tissues
pathology)





AKT
Vaccinate**
none
negative
negative
negative
negative


ALH
Vaccinate**
none
negative
negative
negative
negative


ALU
Vaccinate**
none
negative
negative
negative
negative


ANJ
Vaccinate**
none
negative
negative
negative
negative


ANU
Vaccinate**
none
negative
negative
negative
negative


AJW
Control
none
negative
negative
negative
negative


AKR
Control
none
negative
negative
negative
negative


ALZ
Control
none
negative
negative
negative
negative


ARC
Control
none
negative
negative
negative
negative





*The animals were challenged with an Equine flu isolate Ohio 03


**CARBIGEN ™ adjuvanted inactivated equine flu virus Ohio 03 vaccine was used for vaccination






Example 19—Efficacy of an Equine Influenza Virus Vaccine for Dogs

The canine influenza virus isolated from flu outbreaks in Florida was characterized is closely related to a number of H3N8 type equine influenza virus isolates. By DNA and amino acid sequence similarity analysis it was demonstrated that the canine influenza virus is very similar to an equine influenza virus, A/equine/Ohio/03. The following study was conducted in dogs to determine the efficacy of commercially available equine influenza vaccines in dogs.


Procedure:


Approximately 16 month old, 20 mongrels and 20 beagles of mixed sex were used in the study. The dogs were randomly assigned to 6 groups (Table 31) of 6-7 dogs each. Dogs in groups 1 and 4 were vaccinated with a commercially available inactivated, adjuvanted equine influenza vaccine (EQUICINE™, Intervet Inc., Millsboro, DE) at 16 and 17 months of age via subcutaneous (SQ) route. The dogs in groups 2 and 5 were vaccinated with a modified live equine/Kentucky/91 influenza vaccine in a 1 ml volume via intranasal route (single nostril). Blood samples were collected on the day of vaccination, day 7 and 14 post first vaccination (groups 1, 2, 4, and 5) and post second vaccination (groups 1 and 4) for determining the HI titer using an H3N8 equine influenza virus and a canine influenza virus using per a standard protocol (SAM 124, CVB, USDA, Ames, IA).


Vaccinates (at 72 days post final vaccination) and the controls were challenged oronasally with a cell-culture grown equine influenza virus strain A/equine/Ohio/03 (107.0 to 108.0 TCID50 per dog) in a 1-2 ml volume. The challenge virus was administered to the dogs as mist using a nebulizer (DeVilbiss ULTRA-NEB 99 ultrasonic nebulizer, Sunrise Medical, USA). The dogs were observed for influenza-related clinical signs for 12 days post-challenge. The nasal and oropharyngeal swabs were collected in Earl's MEM medium with antibiotics (neomycin and polymyxin B) daily from day −1 to day 12 post challenge for virus isolation. The presence of virus in the swabs indicates that the animal is excreting the virus in nasal/oral secretions. All dogs were humanely euthanized on day 12 post-challenge and lung tissues were collected in 10% buffered formalin for histopathological evaluation.









TABLE 31







Experimental design













Number


Number
Route of


Group
of dogs
Breed
Treatment
of doses
vaccination





1
7
Beagle
EQUICINE II ™**
2
Subcutaneous


2
7
Beagle
A/KY/91***
1
Intranasal


3
6
Beagle
Control
N/A*
N/A*


4
7
Mongrel
EQUICINE II ™
2
Subcutaneous


5
7
Mongrel
A/KY/91
1
Intranasal


6
6
Mongrel
Control
N/A*
N/A*





*Not applicable


**EQUICINE II ™ is marketed by Intervet Inc. as a liquid vaccine. EQUICINE II ™ contains inactivated A/Pennsylvania/63 influenza (or “A/Pa/63”) virus and A/equine/Kentucky/93 influenza (or “A/KY/93”) virus with carbopol (i.e., HAVLOGEN ® (Intervet Inc.)). More specifically, a dose of EQUICINE II ™ contains: inactivated A/Pa/63 at 106.0 EID50, inactivated A/KY/93 at 106.7 EID50, 0.25% by volume carbopol, and sufficient PBS to create a total volume of 1 ml.


***A/KY/91 is a freeze-dried vaccine that was reconstituted with water. Such reconstitution was conducted using vaccine-grade water sufficient to bring the vaccine dosage to a total volume of 1 ml. The vaccine contained equine/Kentucky/91 influenza (or “A/KY/91”) virus, and is discussed in, for example, U.S. Pat. Nos. 6,436,408; 6,398,774; and 6,177,082, which are incorporated by reference in their entirety into this patent. When reconstituted, a dose of the vaccine contained A/KY/91 at 107.2 TCID50 per ml, 0.015 grams N-Z AMINE AS ™ per ml, 0.0025 grams gelatin per ml, and 0.04 grams D lactose per ml. N-Z AMINE AS ™ is a refined source of amino acids and peptides produced by enzymatic hydrolysis of casein. N-Z AMINE AS ™ is marketed by Kerry Bio-Science (Norwich, NY, USA).







Results:


All vaccinated dogs seroconverted following the vaccination and the HI titers ranged from 10 to 80 for EQUICINE II™ vaccine group dogs compared to 10 to 40 for the A/KY/91 vaccine group dogs using an equine influenza virus (H3N8 type).


The samples collected at 2 weeks post vaccination (post second vaccination for EQUICINE II™ vaccine) were analyzed for HI titer determination with a canine influenza as well as with an equine influenza virus (H3N8 type). The HI results are shown in Table 32. The clinical signs include fever (>103° F.; >39.4° C.), occasional cough, and mild nasal discharge observed following the challenge.









TABLE 32







Serology - HI titers at 2 weeks post vaccination















HI titer with













Number


Equine influenza virus
Canine influenza virus














Group
of dogs
Breed
Treatment
Range
GMT
Range
GMT

















1
7
Beagle
Equicine II ™
10-80
36
10-80
33


2
7
Beagle
A/KY/91
10-20
12
 20-160
54


3
6
Beagle
Control
N/A*
N/A*
N/A*
N/A*


4
7
Mongrel
Equicine II ™
40-80
54
40-80
50


5
7
Mongrel
A/KY/91
10-40
24
40-80
49


6
6
Mongrel
Control
N/A*
N/A*
N/A*
N/A*





*Not applicable






Among beagles, 2 of 6 dogs in the EQUICINE II™ vaccine group (Group 1), 1 of 7 dogs in the A/KY/91 vaccine group (Group 2) and 2 of 6 dogs in the control group (Group 3) had fever. One of 6 dogs in Group 3 (control) was positive for virus in the cell culture supernatant of nasal swab material by hemagglutination assay with 0.25% chicken red blood cells (CRBC). One of 6 dogs in the control group (Group 3) and 1 of 7 dogs in the A/KY/91 vaccine group (Group 2) had mild nasal discharge during the post challenge observation period. There was no statistical significant difference (P >0.05) between control and vaccine groups for beagle dogs.


Among mongrels, 5 of 7 dogs in the EQUICINE II™ vaccine group (Group 4), 1 of 7 dogs in the A/KY/91 vaccine group (Group 5) and 5 of 6 dogs in the control group (Group 6) had fever. One dog from each of Group 4 and 6 had a mild nasal discharge, and one dog from Group 5 had an occasional cough. Two of 7 dogs in the EQUICINE II™ vaccine group (Group 4) and 3 of 6 dogs in the control group (Group 6) were positive for influenza virus in the nasal swab by HA assay. None of the dogs from the A/KY/91 group (Group 5) were positive for influenza virus in the nasal swab materials.


Conclusion:


By serology, it was demonstrated that vaccination of dogs with commercially available equine influenza vaccines stimulated a moderate level influenza antibody response. There may be some breed difference in development of influenza-related clinical signs in dogs following a challenge with H3N8 type influenza virus. The live attenuated equine influenza vaccine (A/KY/91) provided a significant (P<0.05) protection from clinical disease development in rectal temperature in mongrels. Also, the live attenuated viral vaccine prevented the shedding of influenza virus in the nasal secretions.


Example 20-Canine Influenza Challenge Model Development

In view of reports that inducing disease in canines for purposes of study had not proven successful, the potential for using a canine influenza virus, H3N8, to develop a canine influenza challenge model in dogs was investigated in the following study.


Procedure:


Ten mongrels of mixed sex were obtained from a commercial supplier, and housed in cages in a BSL-2 facility. The dogs were randomly assigned to two groups of 5 dogs each. As shown in Table 33, one group was subjected to an intratracheal/intranasal challenge, and the other group was subjected.









TABLE 33







Experimental design











Group
Number of dogs
Challenge route







1
5
Intratracheal/intranasal



2
5
Oronasal











The dogs were challenged at approximately 12 weeks-of-age. Embryonated-chicken-egg grown canine influenza virus (A/canine/Florida/242/03) virus was used as challenge virus. Each dog received a total of approximately 107.2 TCID50 of virus in either 2 ml (for oronasal route) or 4 ml (intratracheal/intranasal route) volume.


For intratracheal/intranasal challenge, 3 ml of the challenge virus was administered into the trachea first, followed by 5 ml of PBS using a delivery tube, which consisted of a cuffed tracheal tube (Size 4.5/5.0, Sheridan, USA) and feeding tube (size 5 Fr, 1.7 mm; 16 inches (41 cm) in length, Kendall, USA), and a 1 ml challenge virus, followed by 3 ml of atmospheric air was administered into nostrils using a syringe.


For oronasal challenge, the challenge virus was administered as a mist using a nebulizer (NEBULAIR™, DVM Pharmaceuticals, Inc., Miami, FL) in approximately 2 ml volume. The dogs were observed for flu-related clinical signs for 14 days post-challenge. The dogs were euthanized at day 14 post challenge, and tissue (lung and trachea) samples were collected in 10% buffered formalin for histopathological examination.


Results:


All dogs in groups 1 and 2 developed canine influenza clinical signs within 24 to 48 hours. Each dog had 2 or more of the following clinical signs: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, weight loss, gagging, hemoptysis, and audible rales. Lung tissues from 5 of 5 dogs from group 1 and 4 of 5 dogs from group 2 had histopathological lesions which included one or more of the following: diffuse suppurative bronchopneumonia, bronchitis/bronchoiolitis with plugs of neutrophilic exudate in the lumina and marked mononuclear cell aggregation in mucosa and peribronchiolar tissue, mixed exudate within alveoli with large numbers of foamy macrophages, lymphocellular and plasma cellular as well as granulocytic cell infiltration, and thickening of alveolar septa with proliferation of type II pneumocytes compatible with or pathognomic to an influenza virus infection. The trachea tissue samples were normal.


Conclusion:


An H3N8 canine influenza isolate such as the one used in this study may be used for inducing canine influenza disease in dogs using one of the methods described in this study or a similar method.


Example 21-Canine Influenza Challenge Model Development

The potential for using a canine influenza virus, H3N8, to develop a canine influenza challenge model in dogs was further investigated in the following study.


Procedure:


Fifteen 17- to 18-week-old mongrels and five 15-week-old beagles were obtained from commercial suppliers, and were housed in cages in a BSL-2 facility. The mongrels were randomly assigned to 3 groups (Groups 1 to 3) of 5 dogs each. All beagles were assigned to one group (Group 4) as shown in Table 34:









TABLE 34







Experimental design












Group
Breed
Number of dogs
Challenge virus dose







1
Mongrels
5
106.8 TCID50



2
Mongrels
5
105.8 TCID50



3
Mongrels
5
104.8 TCID50



4
Beagles
5
106.8 TCID50











The dogs were challenged oronasally with a virulent canine influenza virus, A/Canine/Florida/242/2003 (isolated from lung of a greyhound dog with canine influenza disease (provided by Dr. Cynda Crawford at the University of Florida)). The challenge virus was administered as a mist using a nebulizer (NEBULAIR™ in approximately 2 ml volume. The dogs were observed for flu-related clinical signs for 14 days post-challenge.


Results:


Eighty percent (4 of 5) of the dogs in Group 1 and 4, 100% of the dogs in Group 2 and 3, developed canine influenza clinical signs within 48 hours. Each dog had one or more of the following clinical signs: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, weight loss, gagging, and rales. The clinical signs observed in beagles were generally milder and short-course compared to mongrels.


Conclusion:


An H3N8 canine influenza isolate such as the one used in this study may be used for inducing canine-influenza-like or kennel-cough-like disease in dogs using method described in this study or a similar method with a challenge dose range from 104.8 to 106.8 TCID50. There were some differences in clinical signs observed in mongrels and beagles. In general, beagles tend to have milder flu-related clinical signs compared to mongrels.


Example 22-Canine Influenza Vaccine Efficacy Study

The following study was conducted to assess the efficacy of an H3N8 equine influenza vaccine in dogs against canine influenza virus.


Procedure:


Seventeen 14-week-old mongrels and ten 8-week-old beagles were obtained from commercial suppliers. The dogs were randomly assigned to 5 groups as shown in Table 35, and housed in a research facility.









TABLE 35







Experimental design

















Age at






Number of
Vaccination


Group
Age
Number of dogs
Treatment
doses
(weeks)





1
14 weeks
7
Vaccinate
2
14 & 18


2
14 weeks
5
Vaccinate
1
18


3
14 weeks
5
Control




4
 8 weeks
5
Vaccinate
2
 8 & 12


5
 8 weeks
5
Control











The vaccine used in this study was a HAVLOGEN®-adjuvanted, inactivated equine influenza virus (A/equine/KY/02) vaccine. To prepare this vaccine, the virus was inactivated by binary ethylenimine (BEI) using a standard method. Each vaccine dose contained HAVLOGEN® (10% v/v), 6144 HA units of the inactivated virus, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red, sufficient NaOH to adjust the pH to from 6.8 to 7.2, and sufficient PBS to bring the total dose volume to 1 ml.


The dogs in Groups 1 and 4 were vaccinated with 2 doses of the vaccine. The second dose (i.e., the booster) was administered 4 weeks after the first. The dogs in Group 2 were vaccinated with 1 dose at 18 weeks-of-age. Blood samples were collected to assess HI titer using a standard protocol (e.g., SAM 124, CVB, USDA, Ames, IA) with an H3N8 canine influenza isolate on days zero (before vaccination), 7, and 14 post first and second vaccinations. Approximately 5 days before challenge, the dogs were moved to a BSL-2 facility and housed in individual cages.


All vaccinates and age-matched control dogs were challenged oronasally with a virulent canine influenza virus (107.7 TCID50 of A/Canine/Florida/242/2003 per dog) at 2 weeks post second vaccination of Groups 1 and 4 and first vaccination of Group 2. The challenge virus was administered as a mist using a nebulizer (NEBULAIR™ at 2 ml per dog. The dogs were observed for influenza-related clinical signs for 17 days post-challenge. Nasal and oropharygeal swabs were collected in tubes containing 2 ml of virus transport medium for virus isolation from day −1 (i.e., one day before challenge) to day 17 days post-challenge. All dogs were euthanized at day 17 post-challenge and lung and tracheal samples were collected in 10% buffered formalin for histopathology. Blood samples were collected on days 7 and 14 post challenge for HI titer determination. The clinical sign score assignments used for the post challenge observation are shown in Table 36.


Results:


All dogs in 2-dose vaccination groups (Group 1 and 4) developed HI antibody titer responses to the canine influenza virus isolate (Table 37). Following the challenge, approximately a 4-fold increase in titer on day 14 post challenge in all groups indirectly indicated that all dogs were exposed to the challenge virus. All dogs exhibited one or more of the following signs of canine influenza: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, weight loss, and dyspnea. Vaccinates had less severe clinical signs, compared to age-matched controls (Table 38). There was a significant reduction in clinical signs due to the 2-dose vaccination in both 8-week-old (P=0.040) and 14-week-old (P=0.003) dogs (Groups 4 and 1 respectively). In this experiment, one-dose vaccination did not provide a significant (P=0.294) reduction in clinical signs (Group 2)


Virus isolation results are shown in Table 39. Among 14-week-old dogs, canine influenza virus was isolated from swab samples collected from 2 of 7 dogs (29%) from the 2-dose vaccine group (Group 1), 3 of 5 dogs (60%) from the 1-dose vaccine group (Group 2), and 5 of 5 dogs (100%) from the control group (Group 3). Among 8-week-old dogs, the virus was isolated from 1 of 5 dogs (20%) from the 2-dose vaccine group (Group 4), and 4 of 5 dogs (80%) from the control group (Group 5). There was a significant reduction (P=0.003) in the number of dogs positive for canine influenza virus in swab samples due to 2-dose vaccination (Groups 1 and 4) compared to unvaccinated controls (Groups 3 and 5). Although there was a reduction in the number of dogs (60% vs. 100%) positive for canine influenza virus in swab samples between 1-dose vaccine group (Group 2) and the control group (Group 3), the difference was not statistically significant (P=0.222).


Histopathological evaluation of lung and tracheal tissue samples for lesions was conducted to identify lesions compatible with or pathognomic to canine influenza disease. This includes, for example, determination of whether one or more of the following exist: areas with suppurative bronchopneumonia; peribronchitis/peribronchiolitis with mononuclear cell aggregation (lymphocytes, plasma cells); presence of plugs of granulocytic cellular debris in the lumina; hyperplasia of respiratory epithelium; mixed exudate in the alveoli with large amount of granulocytic cells and cell debris; aggregates of (foamy) macrophages, plasma cells, and lymphocytes; and thickening of alveolar septa with proliferation of type II pneumocytes.


Table 40 provides a summary of the extent of lesions in this experiment for the dogs. Among 14-week-old dogs, the lung lesions were less extensive and less severe in 5 of 7 dogs in the 2-dose vaccination group (Group 2), and 4 of 5 dogs in the 1-dose vaccination group (Group 1). All controls dogs (Group 3) had severe and extensive lesions suggestive of no protection. There was no difference in tracheal lesions due to 1- or 2-dose vaccination among 14-week-old dogs. Among 8-week-old dogs, there was no difference in lung lesions between 2-dose vaccinates and control dogs. None of the dogs had any tracheal lesions.


Conclusion:


The results from this study demonstrate that: (1) inactivated H3N8 equine influenza virus can induce canine influenza virus cross reactive HI antibody responses in vaccinated dogs, (2) use of an H3N8 equine influenza virus vaccine can reduce the severity of canine influenza virus disease in dogs, and (3) use of an H3N8 equine influenza virus vaccine can reduce virus excretion in nasal and/or oral secretions.









TABLE 36







Clinical signs and scoring system










Clinical signs
Score per day







Temp




<103.0° F. (39.4° C.)
0



103.0-103.9° F. (39.4-
2



104.0-104.9° F. (40.0-40.5° C.)
3



>105.0° F. (>40.6° C.)
4



Coughing




No coughing
0



Occasional
2



Paroxysmal
4



Sneezing




No sneezing
0



Occasional
1



Paroxysmal
2



Nasal discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Ocular discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Hemoptysis




No
0



Yes
5



Depression




No
0



Yes
1



Anorexia




No
0



Yes
1



Respiratory signs




None
0



Rales
3



Dyspnea
4



Gasping
5



Mucous expectorate




No
0



Yes
2



Vomiting




No
0



Yes
1



Fecal abnormalities




No
0



Yes
1

















TABLE 37







Serology-Hemagglutination inhibition titers

















HI titer













Group

Age

Number of
Days post first vaccination of Groups 1 and 4
Days post challenge



















No
Dog ID
(week)
Treatment
doses
0*
7
14
28**
35
42***
7
14






















1
921
14
Vaccinate
2
<10
<10
10
20
40
20
160
320


1
926
14
Vaccinate
2
<10
<10
<10
40
40
80
80
>640


1
931
14
Vaccinate
2
<10
<10
<10
10
20
20
80
>640


1
955
14
Vaccinate
2
<10
<10
<10
10
40
40
160
320


1
011
14
Vaccinate
2
<10
<10
<10
10
20
40
160
320


1
013
14
Vaccinate
2
<10
<10
<10
20
40
40
160
320


1
019
14
Vaccinate
2
<10
<10
<10
10
20
40
80
>640


2
922
14
Vaccinate
1
<10
<10
<10
<10
<10
<10
>640
>640


2
953
14
Vaccinate
1
<10
<10
<10
<10
<10
<10
320
>640


2
015
14
Vaccinate
1
<10
<10
<10
<10
<10
<10
320
>640


2
016
14
Vaccinate
1
<10
<10
<10
<10
<10
<10
160
320


2
017
14
Vaccinate
1
<10
<10
<10
<10
<10
<10
320
>640


3
923
14
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


3
012
14
Control
N/A
<10
<10
<10
<10
<10
<10
40
320


3
014
14
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


3
018
14
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


3
01A
14
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


4
406
8
Vaccinate
2
<10
<10
10
40
80
80
160
>640


4
407
8
Vaccinate
2
<10
20
20
40
40
40
320
>640


4
504
8
Vaccinate
2
<10
<10
10
20
20
80
160
>640


4
704
8
Vaccinate
2
<10
<10
10
40
80
160
160
>640


4
705
8
Vaccinate
2
<10
<10
<10
40
80
160
160
>640


5
404
8
Control
N/A
<10
<10
<10
<10
<10
<10
80
160


5
405
8
Control
N/A
<10
<10
<10
<10
<10
<10
80
80


5
610
8
Control
N/A
<10
<10
<10
<10
<10
<10
20
40


5
702
8
Control
N/A
<10
<10
<10
<10
<10
<10
80
160


5
703
8
Control
N/A
<10
<10
<10
<10
<10
<10
40
160





*First vaccination-Groups 1 and 4


**Second vaccination-Groups 1 and 4; First vaccination-Group 2


***Day of challenge













TABLE 38







Analysis of total canine influenza disease clinical scores
















Average





Number
Age at first
total





of doses
vaccination of
Score



Group
Treatment
of vaccine
Groups 1 and 4
per dog
P-value*





1
Vaccinate
2
14 weeks
 8.7
0.003







(Group 1 vs. 3)


2
Vaccinate
1
14 weeks
21.8
0.294





(these dogs were

(Group 2 vs. 3)





vaccinated once,







when they were







18 weeks old)




3
Control

14 weeks
25.4






(these dogs







were not







vaccinated)




4
Vaccinate
2
 8 weeks
 2.0
0.040







(Group 4 vs. 5)


5
Control

 8 weeks
 5.4






(these dogs







were not







vaccinated)





*Analyzed using a NPARIWAY procedure of SAS ® Version 8.2 (the vaccine groups were compared using the Wilcoxon rank sum test)













TABLE 39







Virus shedding
















Number of



Group
Dog
Age

vaccine
Days post-challenge






























No
ID
(week)
Treatment
doses
−1
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17

































1
921
14
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
926
14
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
931
14
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
955
14
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
011
14
Vaccinate
2
N
N
P
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N
N


1
013
14
Vaccinate
2
N
N
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N
N


1
019
14
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
922
14
Vaccinate
1
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
953
14
Vaccinate
1
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
015
14
Vaccinate
1
N
N
N
P
N
P
P
N
N
N
N
N
N
N
N
N
N
N
N


2
016
14
Vaccinate
1
N
N
N
P
N
P
P
N
N
N
N
N
N
N
N
N
N
N
N


2
017
14
Vaccinate
1
N
N
N
N
P
P
N
N
N
N
N
N
N
N
N
N
N
N
N


3
923
14
Control
N/A
N
N
N
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


3
012
14
Control
N/A
N
N
N
P
N
P
N
N
N
N
N
N
N
N
N
N
N
N
N


3
014
14
Control
N/A
N
N
P
N
N
P
P
N
N
N
N
N
N
N
N
N
N
N
N


3
018
14
Control
N/A
N
N
N
P
P
P
N
N
N
N
N
N
N
N
N
N
N
N
N


3
01A
14
Control
N/A
N
N
N
P
P
P
P
N
N
N
N
N
N
N
N
N
N
N
N


4
406
8
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


4
407
8
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


4
504
8
Vaccinate
2
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


4
704
8
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


4
705
8
Vaccinate
2
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


5
404
8
Control
N/A
N
N
P
P
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


5
405
8
Control
N/A
N
N
N
P
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


5
610
8
Control
N/A
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


5
702
8
Control
N/A
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


5
703
8
Control
N/A
N
N
N
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N
















TABLE 40







Histopathological evaluation of tissue samples

















Microscopic lesion


Group

Age

Number of
(Histopathology)













No
Dog ID
(week)
Treatment
doses
Lungs
Trachea
















1
921
14
Vaccinate
2
+/−



1
926
14
Vaccinate
2

+/−


1
931
14
Vaccinate
2




1
955
14
Vaccinate
2
+/−



1
011
14
Vaccinate
2
+/−



1
013
14
Vaccinate
2
+/−
+/−


1
019
14
Vaccinate
2
+/−
+/−


2
922
14
Vaccinate
1
+/−



2
953
14
Vaccinate
1
+/−
+/−


2
015
14
Vaccinate
1
+/−
+


2
016
14
Vaccinate
1




2
017
14
Vaccinate
1
+/−
+/−


3
923
14
Control
N/A
+
+/−


3
012
14
Control
N/A
+



3
014
14
Control
N/A
+



3
018
14
Control
N/A
+



3
01A
14
Control
N/A
+
+/−


4
406
8
Vaccinate
2
+/−



4
407
8
Vaccinate
2




4
504
8
Vaccinate
2
+/−



4
704
8
Vaccinate
2




4
705
8
Vaccinate
2




5
404
8
Control
N/A




5
405
8
Control
N/A




5
610
8
Control
N/A
+/−



5
702
8
Control
N/A
+/−



5
703
8
Control
N/A







“+” Severe lesion consistent or pathognomic to an influenza infection


“+/−“ Mild lesion (inconclusive)


“−“ Normal






Example 23—Canine Influenza Vaccine Efficacy Study

The following study was conducted to determine the efficacy of a multivalent H3N8 equine influenza vaccine against canine influenza virus in dogs.


Procedure:


Seventeen 15-week-old beagles were obtained from a commercial supplier. The dogs were randomly assigned to 3 groups as shown in Table 41, and housed in a research facility.









TABLE 41







Experimental design

















Age at






Number
Vaccination



Group
Number of dogs
Treatment
of doses
(weeks)







1
7
Vaccinate
2
15 & 19



2
5
Vaccinate
1
19



3
5
Control












The vaccine used in this study was a HAVLOGEN® adjuvanted, inactivated equine influenza (A/equine/KY/02, A/equine/KY/93, and A/equine/NM/2/93) vaccine. To prepare this vaccine, the viruses were inactivated by binary ethylenimine (BEI) using a standard method. Each vaccine dose contained HAVLOGEN® (10% v/v), 2048 HA units of each of the inactivated virus, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red, sufficient NaOH to adjust the pH to 6.8 to 7.2, and sufficient PBS to bring the total dose volume to 1 ml.


The dogs in Group 1 were vaccinated with 2 doses of the vaccine. The second (i.e., booster) dose was administered 4 weeks after the first dose. The dogs in Group 2 were vaccinated with 1 dose of vaccine at 19 weeks-of-age. Blood samples were collected to assess HI titer using a standard protocol with an H3N8 canine influenza isolate on days zero (before vaccination), 7, and 14 post first and second vaccinations. Seven days before challenge, the dogs were moved to a BSL-2 facility and housed in individual cages.


All vaccinates and age-matched control dogs were challenged oronasally with a virulent canine influenza virus (107.3 TCID50 of A/Canine/Florida/242/2003 per dog) at 2 weeks post second vaccination of Group 1 and first vaccination of Group 2. The challenge virus was administered as a mist using a nebulizer (NEBULAIR™ at 2 ml per dog. The dogs were observed for influenza-related clinical signs for 14 days post challenge. All dogs were euthanized at day 14 post-challenge, and lung and trachea samples were collected in 10% buffered formalin for histopathology. Blood samples were collected on days 7 and 14 post challenge for HI titer determination. The clinical sign score assignments used for the post challenge observation are shown in Table 42.


Results:


All vaccinated dogs developed HI antibody titer responses to the canine influenza virus isolate (Table 43). Following the challenge, approximately a 4 fold increase in HI titer on day 14 post challenge compared to the pre-challenge HI titer in all groups indirectly indicate that all dogs were exposed to the challenge virus. All dogs exhibited signs canine influenza disease with each dog demonstrating one or more of the following clinical signs: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, weight loss, and dyspnea. Vaccinates had less severe clinical signs, compared to age-matched controls (Table 44). There was a significant (P=0.028) reduction in clinical signs due to the 2-dose vaccination in dogs (Group 1). One dose vaccination did not provide a significant (P=0.068) reduction in clinical signs (Group 2).


As in Example 22, histopathological evaluation of lung and tracheal tissue samples for lesions was conducted to identify lesions compatible with or pathognomic to canine influenza disease. Table 45 provides a summary of the extent of lesions in this experiment for the dogs. Among 15-week-old dogs, vaccination of dogs with either 1 dose or 2 doses prevented the lung lesions in all dogs. Four of 5 control dogs (80%) had severe suppurative bronchopneumonia consistent with an influenza disease. One of 7 dogs from the 2-dose vaccine group (Group 1) and 1 of 5 dogs from the control group (Group 3) had mild trachea lesions suggestive of tracheitis which could be attributed to influenza disease.


Conclusion:


The results from this study demonstrate that 1) inactivated H3N8 equine influenza virus can induce canine influenza virus cross reactive HI antibody responses in vaccinated dogs, and 2) Use of a H3N8 equine influenza virus vaccine can reduce the severity of canine influenza virus disease in dogs.









TABLE 42







Clinical signs and scoring system










Clinical signs
Score per day







Temp




<103.0° F. (39.4° C.)
0



103.0-103.9° F. (39.4-
2



104.0-104.9° F. (40.0-
3



>105.0° F. (>40.6° C.)
4



Coughing




No coughing
0



Occasional
2



Paroxysmal
4



Sneezing




No sneezing
0



Occasional
1



Paroxysmal
2



Nasal discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Ocular discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Hemoptysis




No
0



Yes
5



Depression




No
0



Yes
1



Anorexia




No
0



Yes
1



Respiratory signs




None
0



Rales
3



Dyspnea
4



Gasping
5



Mucous expectorate




No
0



Yes
2



Vomiting




No
0



Yes
1



Fecal abnormalities




No
0



Yes
1

















TABLE 43







Serology - Hemagglutination inhibition titers















HI titer












Group


Number of
Days post first vaccination of Group 1
Days post challenge


















No
Dog ID
Treatment
doses
0*
7
14
28**
35
42***
7
14





















1
ALK
Vaccinate
2
<10
<10
20
20
80
40
160
320


1
AMF
Vaccinate
2
<10
<10
10
20
20
40
160
320


1
AKY
Vaccinate
2
<10
20
20
20
40
40
160
80


1
ALC
Vaccinate
2
<10
10
10
10
40
40
160
160


1
ALL
Vaccinate
2
<10
<10
10
10
40
20
160
320


1
ALM
Vaccinate
2
<10
<10
10
20
40
40
80
160


1
AMU
Vaccinate
2
<10
20
40
40
40
40
40
160


2
ALA
Vaccinate
1
<10
<10
<10
<10
<10
10
320
160


2
AMA
Vaccinate
1
<10
<10
<10
<10
<10
20
>640
80


2
APD
Vaccinate
1
<10
<10
<10
<10
<10
10
>640
320


2
APG
Vaccinate
1
<10
<10
<10
<10
<10
10
320
80


2
APT
Vaccinate
1
<10
<10
<10
<10
<10
10
320
320


3
ALT
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


3
AMS
Control
N/A
<10
<10
<10
<10
<10
<10
80
160


3
AKX
Control
N/A
<10
<10
<10
<10
<10
<10
20
80


3
ALX
Control
N/A
<10
<10
<10
<10
<10
<10
80
80


3
AMI
Control
N/A
<10
<10
<10
<10
<10
<10
40
80





*First vaccination-Group 1


**Second vaccination-Group 1; First vaccination-Group 2


***Day of challenge













TABLE 44







Analysis of total canine influenza disease clinical scores















Age at first
Average





Number
vaccination
total Score



Group
Treatment
of doses
of Group 1
per dog
P-value*





1
Vaccinate
2
15 weeks
 6.3
0.028







(Group 1 vs. 3)


2
Vaccinate
1
15 weeks
14.2
0.068





(these dogs were

(Group 2 vs. 3)





vaccinated







once, when







they were 19







weeks old)




3
Control

15 weeks
24.4






(these dogs







were not







vaccinated)





*Analyzed using a NPARIWAY procedure of SAS ® Version 8.2 (the vaccine groups were compared using the Wilcoxon rank sum test)













TABLE 45







Histopathological evaluation of tissue samples















Microscopic lesion


Group


Number
(Histopathology)












No
Dog ID
Treatment
of doses
Lung
Trachea





1
ALK
Vaccinate
2

+/−


1
AMF
Vaccinate
2




1
AKY
Vaccinate
2




1
ALC
Vaccinate
2




1
ALL
Vaccinate
2




1
ALM
Vaccinate
2




1
AMU
Vaccinate
2




2
ALA
Vaccinate
1




2
AMA
Vaccinate
1




2
APD
Vaccinate
1




2
APG
Vaccinate
1




2
APT
Vaccinate
1




3
ALT
Control
N/A
+/−



3
AMS
Control
N/A
+



3
AKX
Control
N/A
+



3
ALX
Control
N/A
+
+/−


3
AMI
Control
N/A







“+” Severe lesion consistent or pathognomic to an influenza infection


“+/−” Mild lesions (inconclusive)


“−” Normal






Example 24-Canine Influenza Vaccine Efficacy Study

The following study was conducted to determine: (1) the efficacy of monovalent versus multivalent H3N8 equine influenza vaccines against canine influenza virus in dogs, and (2) the effect of route of administration on vaccine efficacy.


Procedure:


Thirty 10-week old mongrels were obtained from a commercial supplier. The dogs were randomly assigned to 6 groups as shown in Table 46, and housed in a research facility.









TABLE 46







Experimental design

















Age at





Route of
Number of
Vaccination


Group
Number of dogs
Treatment
vaccination
doses
(weeks)





1
5
VAX-1
IN
2
10 & 14


2
5
VAX-2
SQ
2
10 & 14


3
5
VAX-2
IN
2
10 & 14


4
5
VAX-3
SQ
2
10 & 14


5
5
VAX-3
IN
2
10 & 14


6
5
Control












Three types of vaccines (VAX-1, VAX-2, and VAX-3) were used. The VAX-1 was a HAVLOGEN®-adjuvanted, inactivated equine influenza virus (A/equine/KY/02) monovalent vaccine, and each dose contained HAVLOGEN® (10% v/v), 6144 HA units of the inactivated virus, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red, sufficient NaOH to adjust the pH to 6.8 to 7.2, and sufficient PBS to bring the total dose volume to 1 ml. The VAX-2 was a HAVLOGEN®-adjuvanted, inactivated equine influenza virus (A/equine/KY/02) monovalent vaccine, and each dose of vaccine contained HAVLOGEN® (10% v/v), 4096 HA units of the inactivated virus, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red, sufficient NaOH to adjust the pH to 6.8 to 7.2, and sufficient PBS to bring the total dose volume to 1 ml. The VAX-3 was a HAVLOGEN®-adjuvanted, inactivated equine influenza (A/equine/KY/02, A/equine/KY/93, and A/equine/NM/2/93) multivalent vaccine, and contained HAVLOGEN® (10% v/v), 2048 HA units of inactivated virus per strain, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red, sufficient NaOH to adjust the pH to 6.8 to 7.2, and sufficient PBS to bring the total dose volume to 1 ml. All influenza viruses used for the vaccine formulation were inactivated by binary ethylenimine (BEI) using a standard method.


The vaccines and routes of administration for each group are described in Table 46. All dogs in the vaccinated groups were vaccinated either via the intranasal (IN) or the subcutaneous (SQ) route, and each dog received 2 doses. The second (i.e., booster) dose was administered 4 weeks after the first dose. Blood samples were collected to assess HI titer using a standard protocol with an H3N8 canine influenza isolate on days zero (before vaccination), 7, and 14 post first and second vaccinations. Seven days before challenge, the dogs were moved to a BSL-2 facility and housed in individual cages.


All vaccinates and age-matched control dogs were challenged oronasally with a virulent canine influenza virus (1074 TCID50 of A/Canine/Florida/242/2003 per dog) at 2 weeks post second vaccination. The challenge virus was administered as a mist using a nebulizer (NEBULAIR™ in a 2 ml volume per day. The dogs were observed for influenza-related clinical signs for 14 days post-challenge. Blood samples were collected on days 7 and 14 post challenge for HI titer determination. All dogs were euthanized at day 14 post-challenge, and lung and trachea samples were collected in 10% buffered formalin for histopathology. The clinical sign score assignments used for the post challenge observation are shown in Table 47.


Results:


All dogs vaccinated via the SQ route developed HI antibody titer responses to the canine influenza virus isolate, regardless of the vaccine type (Table 48). None of the dogs from the IN vaccination groups (i.e., Groups 1, 3, and 5) developed HI antibody titer responses to the canine influenza virus isolate, regardless of the vaccine type, during the post vaccination period. There was, however, a 4-fold increase in titer by day 14 post challenge in all dogs indirectly, indicating that all dogs were exposed to the challenge virus (Table 47).


All dogs exhibited one or more of the following clinical signs of canine influenza: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, weight loss, and dyspnea. Vaccinates had less severe clinical signs, compared to age-matched controls (Table 49). There was a significant reduction in clinical signs in dogs vaccinated with VAX-3 via the SQ route (Group 4). In this experiment, IN administration of either VAX-1, VAX-2, or VAX-3 did not provide a significant reduction in clinical signs of canine influenza virus.


As in Examples 22 and 23, histopathological evaluation of lung and tracheal tissue samples for lesions was conducted to identify lesions compatible with or pathognomic to canine influenza disease. Table 50 provides a summary of the extent of lesions in this experiment for the dogs. Five of 5 control dogs (Group 6) had lung lesions consistence with an influenza infection. Two of 5 dogs vaccinated with VAX-2 via the SC route (Group 2) and 3 of 5 dogs vaccinated with VAX-3 via the SC route (Group 4) were free of any influenza-related lung lesions. All the dogs that received the vaccine via the intranasal route, irrespective of the vaccine type, had severe lung lesions consistent with an influenza infection. The trachea lesions observed in this study were very mild.


Conclusion:


The results from this study demonstrate that: (1) inactivated H3N8 equine influenza virus can induce canine influenza virus cross reactive HI antibody responses in dogs vaccinated via the SQ route, (2) intranasal administration of either monovalent (VAX-1 and VAX-2) or multivalent vaccine (VAX-3) was not efficacious in dogs, and (3) subcutaneous administration of multivalent vaccine (VAX-3) provided a significant (P=0.016) reduction in severity of canine influenza virus disease in dogs.









TABLE 47







Clinical signs and scoring system










Clinical signs
Score per day







Temp




<103.0° F. (<39.4° C.)
0



103.0-103.9° F. (39.4-
2



104.0-104.9° F. (40.0-
3



>105.0° F. (>40.6° C.)
4



Coughing




No coughing
0



Occasional
2



Paroxysmal
4



Sneezing




No sneezing
0



Occasional
1



Paroxysmal
2



Nasal discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Ocular discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Hemoptysis




No
0



Yes
5



Depression




No
0



Yes
1



Anorexia




No
0



Yes
1



Respiratory signs




None
0



Rales
3



Dyspnea
4



Gasping
5



Mucous expectorate




No
0



Yes
2



Vomiting




No
0



Yes
1



Fecal abnormalities




No
0



Yes
1

















TABLE 48







Serology - Hemagglutination inhibition titers

















HI titer













Group


Route of
Number of
Days post vaccination
Days post challenge



















No
Dog ID
Treatment
vaccination
doses
0*
7
14
28**
35
42***
7
14






















1
248
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
40


1
501
Vaccinate
IN
2
<10
10
<10
<10
<10
<10
160
160


1
502
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
160


1
469
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
160


1
46A
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
80


2
232
Vaccinate
SQ
2
<10
<10
<10
20
20
40
320
640


2
511
Vaccinate
SQ
2
<10
10
10
20
20
20
160
640


2
514
Vaccinate
SQ
2
<10
<10
40
40
80
40
160
320


2
461
Vaccinate
SQ
2
<10
10
10
20
20
20
>640
>640


2
463
Vaccinate
SQ
2
<10
10
40
80
80
40
80
320


3
246
Vaccinate
IN
2
<10
10
<10
<10
<10
<10
40
40


3
505
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
80


3
506
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
160


3
464
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
80


3
465
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
160


4
23B
Vaccinate
SQ
2
<10
10
10
40
40
20
160
160


4
247
Vaccinate
SQ
2
<10
<10
<10
20
20
20
160
320


4
508
Vaccinate
SQ
2
<10
10
40
40
80
80
320
320


4
512
Vaccinate
SQ
2
<10
<10
20
20
80
80
320
160


4
516
Vaccinate
SQ
2
<10
10
10
20
80
80
160
>640


5
503
Vaccinate
IN
2
<10
10
<10
<10
<10
<10
80
160


5
513
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
80


5
462
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
320


5
466
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
80


5
46B
Vaccinate
IN
2
<10
<10
<10
<10
<10
<10
80
160


6
236
Control

2
<10
<10
<10
<10
<10
<10
80
160


6
504
Control

2
<10
<10
<10
<10
<10
<10
160
160


6
507
Control

2
<10
<10
<10
<10
<10
<10
80
160


6
515
Control

2
<10
<10
<10
<10
<10
<10
80
160


6
468
Control

2
<10
<10
<10
<10
<10
<10
80
160





*First vaccination


**Second vaccination


***Day of challenge













TABLE 49







Analysis of total canine influenza disease clinical scores













Route of
Average total



Group
Treatment
vaccination
Score per dog
P-value*





1
VAX-1
IN
35.2
0.500






(Group 1 vs. 6)


2
VAX-2
SQ
31.0
0.345






(Group 2 vs. 6)


3
VAX-2
IN
39.4
0.631






(Group 3 vs. 6)


4
VAX-3
SQ
13.0
0.016






(Group 4 vs. 6)


5
VAX-3
IN
42.6
0.790






(Group 4 vs. 6)


6
Control

36.8






*Analyzed using a NPARIWAY procedure of SAS ® Version 8.2 (the vaccine groups were compared using the Wilcoxon rank sum test)













TABLE 50







Histopathological evaluation of tissue samples

















Microscopic lesion


Group


Route of
Number
(Histopathology)













No
Dog ID
Treatment
vaccination
of doses
Lung
Trachea





1
248
Vaccinate
IN
2
+



1
501
Vaccinate
IN
2
+



1
502
Vaccinate
IN
2
+



1
469
Vaccinate
IN
2
+
+


1
46A
Vaccinate
IN
2
+
+


2
232
Vaccinate
SQ
2
+



2
511
Vaccinate
SQ
2
+



2
514
Vaccinate
SQ
2




2
461
Vaccinate
SQ
2
+



2
463
Vaccinate
SQ
2




3
246
Vaccinate
IN
2
+



3
505
Vaccinate
IN
2
+



3
506
Vaccinate
IN
2
+
+


3
464
Vaccinate
IN
2
+



3
465
Vaccinate
IN
2
+
+


4
23B
Vaccinate
SQ
2




4
247
Vaccinate
SQ
2
+/−



4
508
Vaccinate
SQ
2




4
512
Vaccinate
SQ
2

+/−


4
516
Vaccinate
SQ
2
+
+


5
503
Vaccinate
IN
2
+
+/−


5
513
Vaccinate
IN
2
+
+


5
462
Vaccinate
IN
2
+
+/−


5
466
Vaccinate
IN
2
+
+


5
46B
Vaccinate
IN
2
+



6
236
Control

2
+



6
504
Control

2
+
+


6
507
Control

2
+
+


6
515
Control

2
+
+/−


6
468
Control

2
+
+





“+” Severe lesion consistent or pathognomic to an influenza infection


“+/−“ Mild lesion (inconclusive)


“−“ Normal






Example 25-Canine Influenza Vaccine Efficacy Study

Canine influenza disease is caused by an H3N8 influenza virus (CIV). CIV is very closely related to equine H3N8 viruses (Crawford et al., 2005) and infects all exposed dogs. Approximately 80% of exposed dogs develop clinical signs. In the following study the efficacy of an inactivated H3N8 equine influenza virus vaccine and a canine influenza virus vaccine were determined.


Procedure:


Thirty-five beagles and five mongrels were used in this study. Beagles were randomly assigned to three groups (Table 51). All mongrels were assigned to control group (Group 3). All dogs were fed with a standard growth diet and water was available as libitum.









TABLE 51







Experimental design
















Age at





Vaccination
Number of
vaccination



Group
Treatment
route
dogs
(weeks)
Challenge





1
VAX-1
1M
15
8 & 12
Yes


2
VAX-2
SC
 5
8 & 12
Yes


3
Control
N/A
20
N/A
Yes










The dogs in Groups 1 and 2 were vaccinated with either VAX-1 or VAX-2 (Table 51). VAX-1 was a HAVLOGEN® adjuvanted, inactivated equine influenza virus (A/equine/KY/02) vaccine. For vaccine preparation, the vaccine virus was inactivated by binary ethylenimine (BEI) using a standard method. Each dose of vaccine contained HAVLOGEN® (10% v/v), 6144 HA units of the inactivated virus, 0.1% (v/v) of 10% thimerosal, 0.1% (v/v) of phenol red and sufficient PBS to bring the total dose volume to 1 ml and sufficient NaOH to adjust the pH to 6.8 to 7.2.


VAX-2 was an inactivated, CARBIGEN™ adjuvanted, canine influenza antigen vaccine (A/canine/Fl/43/2004). The A/canine/Fl/43/2004 was inactivated by binary ethylenimine (“BEI”) using a standard method. Each dose of the vaccine contained 5% by mass CARBIGEN™, approximately 1280 HA units of the inactivated virus, sufficient PBS to bring the total volume of the dose to 1 ml, and sufficient NaOH to adjust the pH to between 7.2 and 7.4. Serum samples were collected from all dogs on the day of first and second vaccination, days 7 and 14 post first and second vaccinations, and at pre-challenge to determine the HI titers using an H3N8 equine influenza virus standard protocol (SAM 124, CVB, USDA, Ames, IA). Seven days before challenge, the dogs were moved to a ABSL-2 facility and housed in individual cages.


All vaccinates and age-matched control dogs were challenged oronasally with virulent canine influenza virus (107.2 TCID50 of A/Canine/Florida/242/2003 per dog) at 2 weeks post second vaccination. The challenge virus was administered as a mist (2 ml/dog) using a nebulizer (NEBULAIR™). The dogs were observed for influenza-related clinical signs for 14 days post-challenge. Nasal and oropharyngeal swabs were collected daily in tubes containing 2 ml of virus transport medium for virus isolation from day −1 (i.e., one day before challenge) through day 14 post-challenge. Blood samples were collected on days 7 and 14 post challenge for HI titer determination. The clinical sign score assignments used for post challenge observation are shown in Table 52.


Results:


All vaccinated dogs (Groups 1 and 2) developed HI antibody titer responses to the canine influenza virus isolate (Table 53). All dogs exhibited one or more of the following signs of canine influenza: fever (>103.0° F.; >39.4° C.), cough, serous or mucopurulent ocular discharge, serous or mucopurulent nasal discharge, vomiting, diarrhea, depression, and anorexia. Vaccinates had less severe clinical signs, compared to age-matched controls (Table 54). There was a significant (P <0.001) reduction in clinical signs in dogs vaccinated with either VAX-1 (Group 1) or VAX-2 (Group 2).


Virus isolation results are shown in Tables 55 and 56. Following a virulent canine influenza virus challenge, the canine influenza virus was isolated from 5 of 15 (33%) dogs from Group 1 (VAX-1), 0 of 5 (0%) dogs from Group 2 (VAX-2) and 17 of 20 (85%) controls (Group 3). Both inactivated equine influenza vaccine (VAX-1) and canine influenza virus (VAX-2) vaccinates demonstrated a significant (P=0.004) reduction in virus shedding in nasal or oral secretions or both (Table 55) compared to controls.


Conclusion:


The results from this study demonstrate that: (1) inactivated H3N8 equine influenza virus and canine influenza virus vaccines can induce canine influenza virus reactive HI antibody responses in vaccinated dogs, (2) use of an H3N8 equine influenza virus or canine influenza virus vaccine can reduce the severity of canine influenza virus disease in dogs, and (3) use of an H3N8 equine influenza virus or canine influenza virus vaccine can reduce virus excretion in nasal and/or oral secretions.









TABLE 52







Clinical signs and scoring system










Clinical signs
Score per day







Temp




<103.0° F. (<39.4° C.)
0



103.0-103.9° F. (39.4-
2



104.0-104.9° F. (40.0-40.5° C.)
3



>105.0° F. (>40.6° C.)
4



Coughing




No coughing
0



Occasional
2



Paroxysmal
4



Sneezing




No sneezing
0



Occasional
1



Paroxysmal
2



Nasal discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Ocular discharge




No discharge
0



Serous -slight
1



Serous -copious
1



Mucopurulent-slight
2



Mucopurulent-copious
3



Hemoptysis




No
0



Yes
5



Depression




No
0



Yes
1



Anorexia




No
0



Yes
1



Respiratory signs




None
0



Rales
3



Dyspnea
4



Gasping
5



Mucous expectorate




No
0



Yes
2



Vomiting




No
0



Yes
1



Fecal abnormalities




No
0



Yes
1

















TABLE 53







Serology - Hemagglutination inhibition titers















HI titer












Group
Dog

Vaccination
Days post vaccination
Days post challenge


















No
ID
Treatment
route
0*
7
14
28**
35
42***
7
14





















1
AYS
Vaccinate
IM
<10
<10
<10
20
40
40
80
≥640


1
AZV
Vaccinate
IM
<10
<10
<10
20
40
40
160
≥640


1
BAD
Vaccinate
IM
<10
<10
<10
40
40
80
80
320


1
BAE
Vaccinate
IM
<10
<10
10
20
20
20
40
320


1
BAH
Vaccinate
IM
<10
<10
10
10
40
40
160
≥640


1
BAJ
Vaccinate
IM
<10
<10
10
20
80
80
40
320


1
BAN
Vaccinate
IM
<10
10
10
20
40
40
40
320


1
BBN
Vaccinate
IM
<10
10
10
20
80
80
40
320


1
BBT
Vaccinate
IM
<10
<10
<10
20
40
40
40
160


1
BBY
Vaccinate
IM
<10
<10
<10
20
80
80
160
≥640


1
BCS
Vaccinate
IM
<10
10
40
40
160
160
160
160


1
BCZ
Vaccinate
IM
<10
10
10
20
80
40
160
160


1
BDP
Vaccinate
IM
<10
<10
<10
20
40
40
80
≥640


1
BEE
Vaccinate
IM
<10
10
20
40
80
80
160
320


1
BEY
Vaccinate
IM
<10
<10
10
10
40
40
160
160


2
AZH
Vaccinate
SC
<10
<10
10
20
80
80
160
160


2
AZT
Vaccinate
SC
<10
<10
10
10
40
80
320
≥640


2
BBC
Vaccinate
SC
<10
<10
20
40
160
160
80
160


2
BCM
Vaccinate
SC
<10
<10
10
20
80
40
80
160


2
BEB
Vaccinate
SC
<10
<10
<10
10
20
40
80
160


3
AYT
Control
N/A
<10
<10
<10
<10
<10
<10
40
320


3
AZJ
Control
N/A
<10
<10
<10
<10
<10
<10
20
160


3
AZL
Control
N/A
<10
<10
<10
<10
<10
<10
40
160


3
AZN
Control
N/A
<10
<10
<10
<10
<10
<10
160
160


3
BAB
Control
N/A
<10
<10
<10
<10
<10
<10
40
320


3
BBD
Control
N/A
<10
<10
<10
<10
<10
<10
320
≥640


3
BBU
Control
N/A
<10
<10
<10
<10
<10
<10
160
160


3
BBZ
Control
N/A
<10
<10
<10
<10
<10
<10
20
160


3
BCC
Control
N/A
<10
<10
<10
<10
<10
<10
40
320


3
BCD
Control
N/A
<10
<10
<10
<10
<10
<10
80
≥640


3
BCG
Control
N/A
<10
<10
<10
<10
<10
<10
40
≥640


3
BCI
Control
N/A
<10
<10
<10
<10
<10
<10
20
320


3
BCL
Control
N/A
<10
<10
<10
<10
<10
<10
80
≥640


3
BCV
Control
N/A
<10
<10
<10
<10
<10
<10
40
320


3
BDU
Control
N/A
<10
<10
<10
<10
<10
<10
80
≥640


3
MFI
Control
N/A
NT
NT
NT
NT
<10
<10
80
320


3
MFJ
Control
N/A
NT
NT
NT
NT
<10
<10
40
320


3
MFK
Control
N/A
NT
NT
NT
NT
<10
<10
80
320


3
MFR
Control
N/A
NT
NT
NT
NT
<10
<10
80
320


3
MFS
Control
N/A
NT
NT
NT
NT
<10
<10
160
≥640





*First vaccination


**Second vaccination


***Day of challenge













TABLE 54







Analysis of total canine influenza disease clinical scores














Average total




Group
Treatment
Score per dog
P-value*







1
VAX-1
 9.1
<0.001






(Group 1 vs. 3)



2
VAX-2
 5.4
<0.001






(Group 2 vs. 3)



3
Control
24.1








*Analyzed using a NPARIWAY procedure of SAS ® Version 9.1 (the vaccine groups were compared using the GLM procedure)













TABLE 55







Post-challenge virus shedding














Percent dogs




Group
Treatment
excreted the virus
P-value*







1
VAX-1
33% (5/15) 
0.004






(Group 1 vs. 3)



2
VAX-2
 0% (0/5) 
0.004






(Group 2 vs. 3)



3
Control
85% (17/20)








*Analyzed using a FREQ procedure of SAS ® (Version 9.1) and P-value associated with Fisher's exact test













TABLE 56







Serology - Hemagglutination inhibition titers











Group


Vaccination
Days post-challenge


























No
Dog ID
Treatment
route
−1
0
1
2
3
4
5
6
7
8
9
10
11
12
13
14





1
AYS
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
AZV
Vaccinate
IM
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


1
BAD
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BAE
Vaccinate
IM
N
N
N
P
P
N
N
N
N
N
N
N
N
N
N
N


1
BAH
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BAJ
Vaccinate
IM
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


1
BAN
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BBN
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BBT
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BBY
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BCS
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BCZ
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BDP
Vaccinate
IM
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


1
BEE
Vaccinate
IM
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


1
BEY
Vaccinate
IM
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


2
AZH
Vaccinate
SC
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
AZT
Vaccinate
SC
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
BBC
Vaccinate
SC
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
BCM
Vaccinate
SC
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


2
BEB
Vaccinate
SC
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


3
AYT
Control
N/A
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


3
AZJ
Control
N/A
N
N
N
N
N
P
P
N
N
N
N
N
N
N
N
N


3
AZL
Control
N/A
N
N
N
N
N
N
P
N
N
N
N
N
N
N
N
N


3
AZN
Control
N/A
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


3
BAB
Control
N/A
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


3
BBD
Control
N/A
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


3
BBU
Control
N/A
N
N
N
N
N
N
P
N
N
N
N
N
N
N
N
N


3
BBZ
Control
N/A
N
N
N
P
N
N
P
N
N
N
N
N
N
N
N
N


3
BCC
Control
N/A
N
N
N
P
N
P
N
N
N
N
N
N
N
N
N
N


3
BCD
Control
N/A
N
N
N
N
N
P
P
N
N
N
N
N
N
N
N
N


3
BCG
Control
N/A
N
N
N
P
N
P
N
N
N
N
N
N
N
N
N
N


3
BCI
Control
N/A
N
N
N
P
P
P
N
N
N
N
N
N
N
N
N
N


3
BCL
Control
N/A
N
N
N
P
P
N
P
N
N
N
N
N
N
N
N
N


3
BCV
Control
N/A
N
N
N
P
P
N
N
N
N
N
N
N
N
N
N
N


3
BDU
Control
N/A
N
N
N
P
P
N
N
N
N
N
N
N
N
N
N
N


3
MFI
Control
N/A
N
N
N
P
P
P
P
N
N
N
N
N
N
N
N
N


3
MFJ
Control
N/A
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


3
MFK
Control
N/A
N
N
N
P
N
N
N
N
N
N
N
N
N
N
N
N


3
MFR
Control
N/A
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N
N


3
MFS
Control
N/A
N
N
N
P
P
P
N
N
N
N
N
N
N
N
N
N





N - No virus isolated from oral or nasal swabs


P - Virus isolated from nasal or oral or nasal and oral swabs.













TABLE 57







Hemagglutinin, neuraminidase and nucleoprotein gene amino acid


sequence similarities among influenza viruses










Amino acid
Gene of influenza


Gene (Canine/
sequence
virus used for


Florida/43/2004)
similarity
comparison





Hemagglutinin
88
equine/Algiers/72


HA
90
equine/Sao paulo/6/69


HA
91
equine/Miami/1/63


HA
93
equine/Newmarket/79


HA
94
equine/Kentucky/1/81


HA
95
Equi-2/Ludhiana/87


HA
96
Equine/Alaska/1/91


HA
97
equine/Tennessee/5/86


HA
98
equine/Kentucky/5/02


HA
99
equine/Ohio/1/2003


HA
99
canine/Florida/242/2003


Neuraminidase
88
Eq/Algiers/72


NA
90
equine/Sao Paulo/6/69


NA
91
equine/Miami/1/63


NA
93
equine/Newmarket/79


NA
94
equine/Kentucky/1/81


NA
95
Equi-2/Ludhiana/87


NA
96
equine/Santiago/85


NA
97
equine/Tennessee/5/86


NA
98
equine/Kentucky/5/2002


NA
99
equine/Ohio/1/2003


NA
99
canine/Florida/242/2003


Nucleoprotein (“NP”)
94
equi/Miami/1/63


NP
97
equine/Kentucky/1/81


NP
99
equine/Kentucky/5/02


NP
99
equine/Ohio/1/2003


NP
99
canine/Florida/242/2003









The words “comprise,” “comprises,” and “comprising” in this patent (including the claims) are to be interpreted inclusively rather than exclusively. This interpretation is intended to be the same as the interpretation that these words are given under United States patent law.


The above detailed description of preferred embodiments is intended only to acquaint others skilled in the art with the invention, its principles, and its practical application so that others skilled in the art may adapt and apply the invention in its numerous forms, as they may be best suited to the requirements of a particular use. This invention, therefore, is not limited to the above embodiments, and may be variously modified.


It should be understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application.


REFERENCES



  • U.S. Pat. No. 5,106,739

  • U.S. Pat. No. 5,034,322

  • U.S. Pat. No. 6,455,760

  • U.S. Pat. No. 6,696,623

  • U.S. Pat. No. 4,683,202

  • U.S. Pat. No. 4,683,195

  • U.S. Pat. No. 4,800,159

  • U.S. Pat. No. 4,965,188

  • U.S. Pat. No. 5,994,056

  • U.S. Pat. No. 6,814,934

  • U.S. Pat. No. 6,436,408

  • U.S. Pat. No. 6,398,774

  • U.S. Pat. No. 6,177,082

  • Published U.S. Application No. 20040078841

  • Published U.S. Application No. 20040067506

  • Published U.S. Application No. 20040019934

  • Published U.S. Application No. 20030177536

  • Published U.S. Application No. 20030084486

  • Published U.S. Application No. 20040123349

  • Greyhound Daily News, 1/28/99. National Greyhound Association (NGA), Abilene, Kansas http://www.NGAgreyhounds.com.

  • Personal communication, Dr. William Duggar, veterinarian at Palm Beach Kennel Club, West Palm Beach, Florida

  • Altschul, S. F. et al. (1990) “Basic Local Alignment Search Tool” J. Mol. Biol. 215:402-410.

  • Altschul, S. F. et al. (1997) “Gapped BLAST and PSI-BLAST: A New Generation of Protein Database Search Programs” Nucl. Acids Res. 25:3389-3402.

  • An, G. (1987) “Binary Ti vectors for plant transformation and promoter analysis” Methods Enzymol. 153:292-305.

  • Beltz, G. A., Jacobs, K. A., Eickbush, T. H., Cherbas, P. T., Kafatos, F. C. (1983) “Isolation of multigene families and determination of homologies by filter hybridization methods” Methods of Enzymology, R. Wu, L. Grossman and K. Moldave [eds.] Academic Press, New York 100:266-285.

  • Burleson, F. et al. (1992) Virology: A Laboratory Manual (Academic Press).

  • Byars, N. E., A. C. Allison (1987) “Adjuvant formulation for use in vaccines to elicit both cell-mediated and humoral immunity” Vaccine 5:223-228.

  • Crawford, P. C. et al. (2005) “Transmission of equine influenza virus to dogs” Science 310:482-485.

  • Chang, C. P. et al. (1976) “Influenza virus isolations from dogs during a human epidemic in Taiwan” Int J Zoonoses 3:61-64.

  • Dacso, C. C. et al. (1984) “Sporadic occurrence of zoonotic swine influenza virus infections” J Clin Microbiol 20:833-835.

  • de Boer, H. A., Comstock, L. J., Vasser, M. (1983) “The tac promoter: a functional hybrid derived from the trp and lac promoters” Proc. Natl. Acad. Sci. USA 80(1):21-25.

  • Felsenstein, J. (1989) Cladistics 5:164.

  • Fields et al. (1946) Fields Virology, 3rd ed., Lippincott-Raven publishers.

  • Ford, R. B., Vaden, S. L. (1998) “Canine infectious tracheobronchitis” In Infectious Diseases of the Dog and Cat, 2nd edition, C. E. Greene, editor, W.B. Saunders Co., Philadelphia, PA, pp. 33-38.

  • Fouchier et al., (2000) Journal of Clinical Microbiology 38 (11):4096-4101.

  • Good, X. et al. (1994) “Reduced ethylene synthesis by transgenic tomatoes expressing S-adenosylmethionine hydrolase” Plant Molec. Biol. 26:781-790.

  • Guan, Y. et al. (2004) “H5N1 influenza: a protean pandemic threat” Proc Natl Acad Sci USA 101:8156-8161.

  • Guo, Y. et al. (1992) “Characterization of a new avian-like influenza A virus from horses in China” Virology 188:245-255.

  • Houser, R. E. et al. (1980) “Evidence of prior infection with influenza A/Texas/77 (H3N2) virus in dogs with clinical parainfluenza” Can J Comp Med 44:396-402.

  • Karasin, A. I. et al. (2000) “Isolation and characterization of H4N6 avian influenza viruses from pigs with pneumonia in Canada” J Virol 74:9322-9327.

  • Karlin S. and Altschul, S. F. (1990) “Methods for Assessing the Statistical Significance of Molecular Sequence Features by Using General Scoring Schemes” Proc. Natl. Acad. Sci. USA 87:2264-2268.

  • Karlin S. and Altschul, S. F. (1993) “Applications and Statistics for Multiple High-Scoring Segments in Molecular Sequences” Proc. Natl. Acad. Sci. USA 90:5873-5877.

  • Kawaoka, Y. et al., (1989) “Avian-to-human transmission of the PB1 gene of influenza A viruses in the 1957 and 1968 pandemics” J Virol 63:4603-4608.

  • Keawcharoen, J. et al. (2004) “Avian influenza H5N1 in tigers and leopards” Emerg Infect Dis 10:2189-2191.

  • Kendal, A. P. et al. (1982) In Concepts and Procedures for Laboratory-based Influenza Surveillance. A. P. Kendal, M. S. Pereira, J. J. Skehel, Eds. (U.S. Department of Health and Human Services, Centers for Disease Control and Prevention and Pan-American Health Organization, Atlanta, GA, United States) pp. B17-B35.

  • Kilbourne, E.D. et al. (1975) “Demonstration of antibodies to both hemagglutinin and neuraminidase antigens of H3N2 influenza A virus in domestic dogs” Intervirology 6:315-318.

  • Kimura, K. et al. (1998) “Fatal case of swine influenza virus in an immunocompetent host” Mayo Clin Proc 73:243-245.

  • Klimov, A. I. et al. (1992a) “Sequence changes in the live attenuated, cold-adapted variants of influenza A/Leningrad/134/57 (H2N2) virus” Virology 186:795-797.

  • Klimov A. et al. (1992b) “Subtype H7 influenza viruses: comparative antigenic and molecular analysis of the HA-, M-, and NS-genes” Arch Virol. 122:143-161.

  • Kovacova, A. et al. (2002) “Sequence similarities and evolutionary relationships of influenza virus A hemagglutinins” Virus Genes 24:57-63.

  • Kuiken, T. et al. (2004) “Avian H5N1 influenza in cats” Science 306:241.

  • Kumar, S. et al. (2004) “MEGA3: Integrated software for Molecular Evolutionary Genetics Analysis and sequence alignment” Brief Bioinform 5: 150-163.

  • Lee, L.G. et al. (1993) “Allelic discrimination by nick-translation PCR with fluorogenic probes” Nucleic Acids Res. 21(16):3761-3 766.

  • Lewin, B. (1985) Genes II, John Wiley & Sons, Inc., p. 96.

  • Lipatov, A.S. et al. (2004) “Influenza: emergence and control” J Virol 78:8951-8959.

  • Livak, K. J. et al. (1995) “Oligonucleotides with fluorescent dyes at opposite ends provide a quenched probe system useful for detecting PCR product and nucleic acid hybridization” PCR Methods Appl. 4(6):3 57-3 62.

  • Maniatis, T., E.F. Fritsch, J. Sambrook (1982) “Nuclease Bal31Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.

  • Matrosovich, M. et al. (2000) “Early alterations of the receptor-binding properties of H1, H2, and H3 avian influenza virus hemagglutinins after their introduction into mammals” J Virol 74:8502-8512.

  • Maertzdorf et al., (2004) Clin Microbiol. 42(3):981-986.

  • Merrifield, R. B. (1963) “Solid Phase Peptide Synthesis. I. The Synthesis of a Tetrapeptide” J. Amer. Chem. Soc. 85:2149-2154.

  • Nikitin, A. et al. (1972) “Epidemiological studies of A-Hong Kong-68 virus infection in dogs” Bull World Health Organ 47:471-479.

  • Nobusawa, E. et al. (1991) “Comparison of complete amino acid sequences and receptor-binding properties among 13 serotypes of hemagglutinins of influenza A viruses” Virology 182:475-485.

  • Patriarca, P. A. et al. (1984) “Lack of significant person-to person spread of swine influenza-like virus following fatal infection in an immunocompromised child” Am J Epidemiol 119:152-158.

  • Payungporn S. et al. (2006a) “Detection of canine influenza A virus (H3N8) RNA by real-time RT-PCR” (in preparation for Journal of Clinical Microbiology).

  • Payungporn S, et al. (2006b) “Isolation and characterization of influenza A subtype H3N8 viruses from dogs with respiratory disease in a shelter and veterinary clinic in Florida” (in preparation for Emerging Infectious Diseases).

  • Peiris, M. et al. (1999) “Human infection with influenza H9N2” Lancet 354:916-917.

  • Peiris, J. S. et al. (2004) “Re-emergence of fatal human influenza A subtype H5N1 disease” Lancet 363:617-619.

  • Posnett, D. N. et al. (1988) “A Novel Method for Producing Anti-peptide Antibodies” J. Biol. Chem. 263(4):1719-1725.

  • Putnam, Bob (Feb. 10, 1999) “Two illnesses seen in death of dogs” St. Petersburg Times.

  • Reid, A. H. et al. (2004) “Evidence of an absence: the genetic origins of the 1918 pandemic influenza virus” Nat Rev Microbiol 2:909-914.

  • Rowe, T. et al. (1999) “Detection of antibody to avian influenza A (H5N1) virus in human serum by using a combination of serologic assays” J Clin Microbiol 37: 937-943.

  • Saiki, R. (1985) “Enzymatic amplification of beta-globin genomic sequences and restriction site analysis for diagnosis of sickle cell anemia” Science 230:1350-1354.

  • Sambrook, J. et al. (1989) “Plasmid Vectors” In: Molecular Cloning: A Laboratory Manual, 2d Edition, pp. 1.82-1.104. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.

  • Subbarao, K. et al. (1998) “Characterization of an avian influenza A (H5N1) virus isolated from a child with a fatal respiratory illness” Science 279:393-396.

  • Suzuki, Y. et al. (2000) “Sialic acid species as a determinant of the host range of influenza A viruses” J Virol 74:11825-11831.

  • Tam, J. P. (1988) “Synthetic Peptide Vaccine Design: Synthesis and Properties of a High-Density Multiple Antigenic Peptide System” PNAS USA 85(15):5409-5413.

  • Top, Jr., F. H. et al. (1977) “Swine influenza A at Fort Dix, New Jersey (January-February 1976). IV. Summary and speculation” J Infect Dis 136 Suppl:S376-5380.

  • Vines, A. et al. (1998) “The role of influenza A virus hemagglutinin residues 226 and 228 in receptor specificity and host range restriction” J Virol 72:7626-7631.

  • Wagner, R. et al. (2002) “N-Glycans attached to the stem domain of haemagglutinin efficiently regulate influenza A virus replication” J Gen Virol 83:601-609.

  • Webby, R. et al. (2004) “Molecular constraints to interspecies transmission of viral pathogens” Nat Med 10:S77-S81.

  • Webster, R. G. (1998) “Influenza: an emerging disease” Emerg Infect Dis. 4:436-441.

  • Webster, R. G. et al. (1992) “Evolution and ecology of influenza A viruses” Microbiol Rev. 56:152-179.

  • Weis, W. et al. (1988) “Structure of the influenza virus haemagglutinin complexed with its receptor, sialic acid” Nature 333:426-431.

  • Womble, D. D. (2000) “GCG: The Wisconsin Package of sequence analysis programs” Methods Mol Biol 132:3-22.

  • Xu, D., McElroy, D., Thornburg, R. W., Wu, R. et al. (1993) “Systemic induction of a potato pin2 promoter by wounding, methyl jasmonate, and abscisic acid in transgenic rice plants” Plant Molecular Biology 22:573-588.

  • Yang, T. T. et al. (1996) “Optimized Codon Usage and Chromophore Mutations Provide Enhanced Sensitivity with the Green Fluorescent Protein” Nucleic Acid Research 24(22):4592-4593.

  • Yoon K-Y. et al. (2005) “Influenza virus infection in racing greyhounds” Emerg Infect Dis. 11:1974-1975.


Claims
  • 1. A reassortant virus comprising a donor influenza virus polynucleotide sequence comprising at least one gene, genomic segment, or a polynucleotide encoding a canine influenza virus protein selected from the group consisting of HA, NA, NP, MA, PB1, PB2, PA, and NS, or a functional or immunogenic fragment thereof and comprising a nucleotide sequence selected from the group consisting of SEQ ID NOs: 1, 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, and 77 and a recipient influenza virus genome from a second influenza virus.
  • 2. A composition comprising a reassortant virus according to claim 1 and a pharmaceutically acceptable carrier or diluent.
  • 3. The reassortant virus according to claim 1, wherein said HA protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 16, 32, 62, and 78.
  • 4. The reassortant virus according to claim 1, wherein said NA protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 28, 58, and 74.
  • 5. The reassortant virus according to claim 1, wherein said NP protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 26, 56, and 72.
  • 6. The reassortant virus according to claim 1, wherein said MA protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 14, 30, 60, and 76.
  • 7. The reassortant virus according to claim 1, wherein said PB1 protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 20, 50, and 66.
  • 8. The reassortant virus according to claim 1, wherein said PB2 protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 18, 48, and 64.
  • 9. The reassortant virus according to claim 1, wherein said PA protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 22, 52, and 68.
  • 10. The reassortant virus according to claim 1, wherein said NS protein has an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 24, 54, and 70.
  • 11. The composition according to claim 2, wherein said pharmaceutically acceptable carrier or diluent is water, saline, an oil, ethanol, dimethyl sulfoxide, gelatin, a cyclodextran, magnesium stearate, dextrose, cellulose, a sugar, calcium carbonate, glycerol, alumina, starch, a suspension agent, a protectant, a lubricant, a buffer, a preservative, a stabilizer, or any combination thereof.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. Ser. No. 16/382,429, filed Apr. 12, 2019, now U.S. Pat. No. 11,160,859, which is a divisional of U.S. Ser. No. 15/888,715, filed Feb. 5, 2018, now U.S. Pat. No. 10,258,686, which is a divisional of U.S. Ser. No. 14/507,332, filed Oct. 6, 2014, now U.S. Pat. No. 9,913,892, which is a divisional of U.S. Ser. No. 13/066,651, filed Apr. 19, 2011, now U.S. Pat. No. 9,345,758, which is a divisional of U.S. Ser. No. 11/584,818, filed Oct. 19, 2006, now U.S. Pat. No. 7,959,929, which is a continuation-in-part of U.S. Ser. No. 11/409,416, filed Apr. 21, 2006, now abandoned, which claims priority from U.S. Ser. No. 60/673,443, filed Apr. 21, 2005; and U.S. Ser. No. 11/584,818 claims priority to U.S. Serial Nos. 60/728,449, filed Oct. 19, 2005; 60/754,881, filed Dec. 29, 2005; 60/759,162, filed Jan. 14, 2006; 60/761,451, filed Jan. 23, 2006; and 60/779,080, filed Mar. 3, 2006, the disclosure of each of which is hereby incorporated by reference herein in its entirety, including any brief summary, detailed descriptions of the invention, examples, claims, abstract, figures, tables, nucleic acid sequences, amino acid sequences, and drawings. The Sequence Listing for this application is labeled “2578403.TXT” which was created on Sep. 9, 2021 and is 400 KB. The entire contents of the sequence listing is incorporated herein by reference in its entirety.

US Referenced Citations (83)
Number Name Date Kind
3541206 Hall Nov 1970 A
3655871 Werner Apr 1972 A
3869546 Lund Mar 1975 A
4009258 Kilbourne Feb 1977 A
4024235 Weetall et al. May 1977 A
4206287 Hannoun et al. Jun 1980 A
4500513 Brown et al. Feb 1985 A
4631191 Dale et al. Dec 1986 A
4683137 Coggins et al. Jul 1987 A
4683195 Mullis et al. Jul 1987 A
4683202 Mullis Jul 1987 A
4689224 Bull et al. Aug 1987 A
4800159 Mullis et al. Jan 1989 A
RE33164 Brown et al. Feb 1990 E
4911910 Mifflin Mar 1990 A
4965188 Mullis et al. Oct 1990 A
5034322 Rogers et al. Jul 1991 A
5106739 Comai et al. Apr 1992 A
5674499 Willemse et al. Oct 1997 A
5756103 Paoletti et al. May 1998 A
5783195 Cochran et al. Jul 1998 A
5994056 Higuchi Nov 1999 A
6017542 Paoletti et al. Jan 2000 A
6033886 Conzelmann Mar 2000 A
6045790 Campbell Apr 2000 A
6177082 Dowling et al. Jan 2001 B1
6207166 Audonnet et al. Mar 2001 B1
6309647 Paoletti et al. Oct 2001 B1
6344354 Webster et al. Feb 2002 B1
6348450 Tang et al. Feb 2002 B1
6398774 Penner et al. Jun 2002 B1
6436408 Dowling et al. Aug 2002 B1
6455760 Zhao et al. Sep 2002 B1
6482414 Dowling et al. Nov 2002 B1
6558674 Audonnet et al. May 2003 B1
6579528 Dowling et al. Jun 2003 B1
6696623 Doerner et al. Feb 2004 B1
6713068 Audonnet et al. Mar 2004 B1
6727078 Montelaro et al. Apr 2004 B2
6814934 Higuchi Nov 2004 B1
7029903 Dowling et al. Apr 2006 B2
7033748 Hillman Apr 2006 B2
7037707 Webster et al. May 2006 B2
7041300 Sanchez et al. May 2006 B1
7074414 Dowling et al. Jul 2006 B2
7163926 Audonnet et al. Jan 2007 B1
7244435 Lai Jul 2007 B2
7468187 Yoon et al. Dec 2008 B2
7572620 Olsen et al. Aug 2009 B2
7682619 Dubovi Mar 2010 B2
7959929 Crawford et al. Jun 2011 B2
9345758 Crawford et al. May 2016 B2
9814770 Olsen et al. Nov 2017 B2
9913892 Crawford et al. Mar 2018 B2
20020025325 Chu et al. Feb 2002 A1
20020071848 Smith et al. Jun 2002 A1
20030045492 Tang et al. Mar 2003 A1
20030084486 Bruce et al. May 2003 A1
20030177536 Grundler et al. Sep 2003 A1
20030180322 Dowling et al. Sep 2003 A1
20040019934 Ekramoddoullah et al. Jan 2004 A1
20040029251 Hoffman et al. Feb 2004 A1
20040052775 Sanchez et al. Mar 2004 A1
20040067506 Scheres et al. Apr 2004 A1
20040078841 Atkinson et al. Apr 2004 A1
20040123349 Xie et al. Jun 2004 A1
20040142450 Seo et al. Jul 2004 A1
20040180058 Shneider et al. Sep 2004 A1
20050013826 Shneider et al. Jan 2005 A1
20050054846 Webster et al. Mar 2005 A1
20060057163 Audonnet et al. Mar 2006 A1
20060153871 Olsen et al. Jul 2006 A1
20060286591 Duke et al. Dec 2006 A1
20070009548 Sterner et al. Jan 2007 A1
20070031512 Hughes Feb 2007 A1
20070031941 Duke et al. Feb 2007 A1
20070048819 Minke et al. Mar 2007 A1
20070048821 Minke et al. Mar 2007 A1
20070065452 Schiltz et al. Mar 2007 A1
20070082012 Shields et al. Apr 2007 A1
20070092537 Chiang et al. Apr 2007 A1
20070098742 Yoon et al. May 2007 A1
20070253981 Dubovi Nov 2007 A1
Foreign Referenced Citations (64)
Number Date Country
722699 Mar 1999 AU
1 035 209 Sep 2000 EP
1067463 May 1967 GB
1183506 Mar 1970 GB
1212886 Nov 1970 GB
1450557 Sep 1976 GB
2000-230931 Aug 2000 JP
2002-514885 May 2002 JP
2004-285019 Oct 2004 JP
2003-520573 Aug 2007 JP
2084524 Jul 1997 RU
2193065 Nov 2002 RU
WO 1987001942 Apr 1987 WO
WO 199417826 Aug 1994 WO
WO 1995007994 Mar 1995 WO
WO 9527780 Oct 1995 WO
WO 199617072 Jun 1996 WO
WO 9637624 Nov 1996 WO
WO 199734008 Sep 1997 WO
WO 199803198 Jan 1998 WO
WO 199908713 Feb 1999 WO
WO 199951269 Oct 1999 WO
WO 9957284 Nov 1999 WO
WO 0009702 Feb 2000 WO
WO 0104333 Jan 2001 WO
WO 200103736 Jan 2001 WO
WO 0109291 Feb 2001 WO
WO 2001011081 Feb 2001 WO
WO 200160849 Aug 2001 WO
WO 200162902 Aug 2001 WO
WO 200197792 Dec 2001 WO
WO 200197804 Dec 2001 WO
WO 2002002139 Jan 2002 WO
WO 2002089586 Nov 2002 WO
WO 03061555 Jul 2003 WO
WO 2003068923 Aug 2003 WO
WO 03086453 Oct 2003 WO
WO 2003086454 Oct 2003 WO
WO 03091401 Nov 2003 WO
WO 2004027049 Apr 2004 WO
WO2004057021 Jul 2004 WO
WO 2004058188 Jul 2004 WO
WO 2005002618 Jan 2005 WO
WO 200514778 Feb 2005 WO
WO 2005024039 Mar 2005 WO
WO 2005062820 Jul 2005 WO
WO 2005120564 Dec 2005 WO
WO 2006073436 Jul 2006 WO
WO 2006088481 Aug 2006 WO
WO 2006108846 Oct 2006 WO
WO 2006116082 Nov 2006 WO
WO 2006128294 Dec 2006 WO
WO 2007002007 Jan 2007 WO
WO 2007002008 Jan 2007 WO
WO 2007019250 Feb 2007 WO
WO 2007024947 Mar 2007 WO
WO 2007027321 Mar 2007 WO
WO 2007042884 Apr 2007 WO
WO 2007047728 Apr 2007 WO
WO 2007048086 Apr 2007 WO
WO 2007061969 May 2007 WO
WO 2007118206 Oct 2007 WO
WO 2007124327 Nov 2007 WO
WO 2008076371 Jun 2008 WO
Non-Patent Literature Citations (139)
Entry
“Animal alerts, a scary flu; a move to change laws to save pets.” Belvoir Media Group, LLC, Nov. 22, 2005, http://www.whole-dog-journal.com/issues/8_11/features/15765-1.html.
“Appropriate Samples for Detecting the Presence of Canine Influenza Virus” Cornell University Animal Health Diagnostic Center, Mar. 27, 2006, http://diaglab.vet.cornell.edu/pdf/CIV-Samp.pdf.
“Canine Influenza Backgrounder.” AVMA, Oct. 24, 2005 http://www.avma.org/public_health/influenza/canine_bgnd.asp.
“Canine Influenza Virus Detected in Dogs in New York State” Cornell University Animal Health Diagnostic Center, Sep. 29, 2005, http://diaglab.vet.cornell.edu/pdf/CIV-NYS.pdf.
“European advice on H5N1 avian influenza in cats.” The Veterinary Record, News & Reports, Mar. 11, 2006, p. 314.
Accession No. AY855338, “Influenza A virus (A/equine/Kentucky/5/02(H3N8)) PB2 polymerase 2 gene, complete cds” Mar. 12, 2005.
Altschul, S. F. et al. “Basic Local Alignment Search Tool”, J. Mol. Biol., 1990, pp. 403-410, vol. 215.
Altschul, S. F. et al. “Gapped BLAST and PSI-BLAST: A New Generation of Protein Database Search Programs”, Nucl. Acids Res., 1997, pp. 3389-3402, vol. 25. No. 17.
Butler, D., “Thai dogs carry bird-flu virus, but will they spread it?” Nature, Feb. 16, 2006, pp. 773, vol. 439.
Byars, N. E. et al. “Adjuvant Formulation for Use in Vaccines to Elicit Both Cell-Mediated and Humoral Immunity”, Vaccine, 1987, pp. 223-228, vol. 5.
Carey, S. “Are dogs susceptible for equine influenza virus?” DVM Newsmagazine, Jun. 1, 2004.
Carey, S. “UF Researchers: Equine Influenza Virus Likely Involved in Recent Respiratory Disease Outbreak in Racing Greyhounds”, HSC News and Communications, University of Florida, Apr. 22, 2004.
Castleman, W., et al. “Pathologic findings in dogs infected with newly emerged canine H3N8 influenza virus.” FASEB Journal, Mar. 6, 2006, pp. A214, vol. 20, No. 4. Abstract Only.
Cattoli, G. et al. “Plague Morphology of Italian H7N1 LPAI isolates in MDCK cells and in primary cells of different avian species” Avian Diseases, 2003, 47:1161-1163.
CDC Media Relations—Telebriefing Transcript—Sep. 26, 2005, http://www.cdc.gov/od/oc/media/transcripts/t050926.htm.
CDC Media Relations “Media Briefing on Canine Influenza” CDC Media Relations—Telebriefing Transcript, Sep. 26, 2005, pp. 1-13, www.cdc.gov/od/oc/media/transcripts/t050926.htm.
Claas, E.C.J. et al. “Human influenza A H5N1 virus related to a highly pathogenic avian influenza virus” The Lancet, 1998, 351:472-477.
Clark, A., “Canine Influenza Virus Surfaces.” JAVMA, Nov. 1, 2005, pp. 1377-1378, vol. 227, No. 9.
Crawford, P. C. et al. “Supporting Online Material forTransmission of Equine Influenza Virus to Dogs” Science, Sep. 29, 2005, pp. S1-S19, http://www.sciencemag.org/cgi/content/full/1117950/DC1.
Crawford, P. C. et al. “Transmission of Equine Influenza Virus to Dogs”, Science, 2005, pp. 482-485, vol. 310.
Dacso, C. C. et al. “Sporadic occurrence of Zoonotic Swine Influenza Virus Infections”, J. Clin. Microbiol., 1984, pp. 833-835, vol. 20, No. 4.
Daly, J.M., “Equine influenza in dogs: Too late to bolt the stable door?” The Veterinary Journal, 2006, pp. 7-8, vol. 171.
Database Biosis, Accession No. PREV200300495526, Cattoli, G. et al., “Plaque morphology of Italian H7N1 LPAI isolates in MDCK cells and in primary cells of different avian species” Biosciences Information Service, 2003.
Database EMBL, Accession No. Q30BG2, “Matrix Protein 1,” Oct. 31, 2006, 1 page.
Database EMBL, Accession No. Q3I3U6, “M1”, Nov. 8, 2005, pp. 1-2.
Database Medline, Accession No. NLM15782624, Emanuela, F. et al., “Detection of influenza A virus by RT-PCR and standard methods in experimental infection of ducks” US National Library of Medicine, Jan. 2005.
De Boer, H. A. et al. “The Tac Promoter: A Functional Hybrid Derived from the trp and lac Promoters”, Proc. Natl. Acad. Sci. USA, 1983, pp. 21-25, vol. 80, No. 1.
De Wit et al., “Efficient generation and growth of influenza virus A/PR/8/34 from eight cDNA fragments”, Virus Research, 2004, pp. 155-161, vol. 103.
Dubovi, E.J., Crawford, C.W., Donis, R.O., Castleman, W.L., Stephenson, I., Gibbs, E.P.J. “Isolation of equine influenza virus from racing greyhounds with fatal hemorrhagic pneumonia” In: Proceedings of the 47th Annual Meeting of American Association of Veterinary Laboratory Diagnosticians, Oct. 2004, Greensboro, North Carolina, p. 158.
Edinboro CH et al., “A placebo-controlled trial of two intranasal vaccines to prevent tracheobronchitis (kennel cough) in dogs entering a humane shelter” Preventative Veterinary Medicine, 2004, 62:89-99.
Foni et al. “Detection of influenza A virus by RT-PCR and standard methods in experimental infection of ducks” The New Microbiologica, 2005, 28:31-35.
Fouchier et al. “Detection of Influenza A Viruses from Different Species by PCR Amplification of Conserved Sequences in the Matrix Gene” Journal of Clinical Microbiology, 2000, pp. 4096-4101, vol. 38, No. 11.
Fouchier, R.A.M. et al. “Characterization of a novel influenza A virus hemagglutinin subtype (H16) obtained from black-headed gulls” J. Virology, 2005, 79(5):2814-2822.
Genbank Accession #AAX23575, hemagglutinin precursor [Influenza A virus (A/equine/Kentucky/5/2002(H3N8))], Mar. 12, 2005.
GenBank Accession AY855341.1 “Influenza A virus (A/equine/Kentucky/5/02(H3N8)) hemagglutinin precursor” Mar. 12, 2005.
GenBank Accession AY919314.1 Influenza A virus (A/Equi 2/Gotland/01 (H3N8)) Mar. 1, 2005.
GenBank Accession No. ABA41538, hemagglutinin [Influenza A virus (A/canine/Iowa/13628/2005(H3N8))], 2005.
GenBank Accession No. DQ124196, “Influenza A virus (A/canine/Texas/1/2004(H3N8)) hemagglutinin gene, partial cds.” Jan. 25, 2006.
GenBank Accession No. DQ146419, Influenza A virus (A/canine/Iowa/13628/2005(H3N8)) hemagglutinin (HA) gene, complete cds, 2005.
Gibbs, E.P.J., “Emerging zoonotic epidemics in the interconnected global community.” The Veterinary Record, Nov. 26, 2005, pp. 673-679, vol. 157.
Good, X. et al. “Reduced Ethylene Synthesis by Transgenic Tomatoes Expressing S-Adenosylmethionine Hydrolase”, Plant Molec. Biol., 1994, pp. 781-790, vol. 26.
Greyhound Daily News, Jan. 28, 1999. National Greyhound Association (NGA), Abilene, Kansas. http://www.NGAgreyhounds.com.
Guan, Y. et al. “H5N1 Influenza: A Protean Pandemic Threat”, Proc. Natl Acad. Sci. USA, 2004, pp. 8156-8161, vol. 101, No. 21.
Guo, Y et al. “Characterization of a New Avian-Like Influenza A Virus from Horses in China”, Virology, 1992, pp. 245-255, vol. 188, No. 1. Abstract Only.
Hampton, T., “Equine Influenza Jumps to Canines.” JAMA, Oct. 26, 2005 pp. 2015, vol. 294, No. 16.
Hanshaw, D., et al. “Canine H3N8 influenza virus infection in mice.” FASEB Journal, Apr. 28, 2007, pp. A406, vol. 21, No. 5. Abstract Only.
Houser, R. E. et al. “Evidence of Prior Infection with Influenza A/Texas/77 (H3N2) Virus in Dogs with Clinical Parainfluenza”, Can. J. Comp. Med., Oct. 1980, pp. 396-402, vol. 44.
IBIS—Integrated Biotechnological Information Services, Query: SA693587:78, Subject: UNIPROT:Q3I3Q4, Sequence Alignment Relating to Crawford et al., “Transmission of equine influenza virus to dogs,” Science, 2005, vol. 310, pp. 482-485.
IBIS—Integrated Biotechnological Information Services, Query: SA693587:78, Subject: GSP:AFU64065, Sequence Alignment relating to WO 2007/048086, published on Apr. 26, 2007.
Jeremijenko, A., et al., “From the front lines.” Nature, Apr. 6, 2006, pp. 726-727, vol. 440.
Karaca, K. et al. “Evaluation of the Ability of Canarypox-Vectored Equine Influenza Virus Vaccones to Induce Humoral Immune Responses against Canine Influenza Viruses in Dogs”, AJVR, Feb. 2007, pp. 208-212, vol. 68, No. 2.
Karaca, K., et al. “Immunogenicity of Fowlpox Virus Expressing the Avian Influenza Virus H5 Gene (TROVAC AIV-H5) in Cats.” Clinical and Diagnostic Laboratory Immunology, Nov. 2005, pp. 1340-1342, vol. 12, No. 11.
Karasin, A. I. et al. “Isolation and Characterization of H4N6 Avian Influenza Viruses from Pigs with Pneumonia in Canada”, J. Virol., 2000, pp. 9322-9327, vol. 74, No. 19.
Karlin, S. et al. “Applications and Statistics for Multiple High-Scoring Segments in Molecular Sequences”, Proc. Natl. Acad. Sci. USA, 1993, pp. 5873-5877, vol. 90.
Karlin, S. et al. “Methods for Assessing the Statistical Significance of Molecular Sequence Features by Using General Scoring Schemes”, Proc. Natl. Acad. Sci. USA, 1990, pp. 2264-2268, vol. 87.
Kawaoka, Y. et al “Avian-to-Human Transmission ofthe PB1 Gene of Influenza A Viruses in the 1957 and 1968 Pandemics”, J. Virol., 1989, pp. 4603-4608, vol. 63, No. 11.
Keawcharoen, J. et al. “Avian Influenza H5N1 in Tigers and Leopards”, Emerg. Infect. Dis., 2004, pp. 2189-2191, vol. 10, No. 12.
Kilbourne, E. D. et al. “Demonstration of Antibodies to Both Hemagglutinin and Neuraminidase Antigens of H3N2 Influenza A Virus in Domestic Dogs”, Intervirology, 1975, pp. 315-318, vol. 6.
Kimura, K. et al. “Fatal Case of Swine Influenza Virus in an Immunocompetent Host”, Mayo Clin. Proc., 1998, pp. 243-245, vol. 73.
Klimov, A. et al. “Subtype H7 Influenza Viruses: Comparative Antigenic and Molecular Analysis of the HA-, M-, and NS-genes”, Arch Virol., 1992, pp. 143-161, vol. 122.
Klimov, A. I. et al. “Sequence Changes in the Live Attenuated, Cold-Adapted Variants of Influenza A/Leningrad/134/57 (H2N2) Virus”, Virology, 1992, pp. 795-797, vol. 186, No. 2. Abstract Only.
Kovacova, A. et al. “Sequence Similarities and Evolutionary Relationships of Influenza Virus A Hemagglutinins”, Virus Genes, 2002, pp. 57-63, vol. 24, No. 1.
Kuiken, T. et al. “Avian H5N1 Influenza in Cats”, Science, 2004, pp. 241, vol. 306.
Kuiken, T., et al. “Feline friend or potential foe?” Nature, Apr. 6, 2006, pp. 741-742, vol. 440.
Lai, A. C. K. et al. “Alternate Circulation of Recent Equine-2 Influenza Viruses (H3N8) from Two Distinct Lineages in the United States”, Virus Research, 2004, pp. 159-164, vol. 100.
Lee, L. G. et al. “Allelic Discrimination by Nick-Translation PCR with Fluorogenic Probes”, Nucleic Acids Res., 1993, pp. 3761-3766, vol. 21, No. 16. Abstract Only.
Lewis, D.B., “Avian Flu to Human Influenza.” Annu. Rev. Med. 2006, pp. 139-154, vol. 57.
Lin, Y.P. et al. “Avian-to-human transmission of H9N2 subtype influenza A viruses: Relationship between H9N2 and H5N1 human isolates” PNAS, 2000, 97(17):9654-9658.
Lipatov, A. S. et al. “Influenza: Emergence and Control”, J. Virol., 2004, pp. 8951-8959, vol. 78, No. 17.
Livak, K. J. et al. “Oligonucleotides with Fluorescent Dyes at Opposite Ends Provide a Quenched Probe System Useful for Detecting PCR Product and Nucleic Acid Hybridization”, PCR Methods Appl., 1995, pp. 357-362, vol. 4, No. 6. Abstract Only.
Maertzdorf et al. “Real-Time Reverse Transcriptase PCR Asay for Detection of Human Metapneumonoviruses from All Known Genetic Lineages”, Clin. Microbiol., 2004, pp. 981-986, vol. 42, No. 3.
Manuguerra, J. C. et al. “Natural infection of dogs by influenza C virus” Res. Virol., 1992, pp. 199-204, vol. 143.
Massin, P. et al., “Cloning of the Chicken RNA Polymerase I Promoter and Use for Reverse Genetics of Influenza A Viruses in Avian Cells”, Journal of Virology, Nov. 2005, p. 13811-13816.
Matrosovich, M. et al. “Early Alterations of the Receptor-Binding Properties of H1, H2, and H3 Avian Influenza Virus Hemagglutinins After Their Introduction into Mammals”, J. Virol., 2000, pp. 8502-8512, vol. 74, No. 18.
McNeil, D.G. and Rubenstein, C. “A New Deadly, Contagious Dog Flu Virus is Detected in 7 States”, The New York Times (nytimes.com), Sep. 22, 2005.
Merrifield, R. B. et al. “Solid Phase Peptide Synthesis. I. The Synthesis of a Tetrapeptide”, J. Amer. Chem. Soc., 1963, pp. 2149-2154, vol. 85.
Mochalova et al., “Receptor-binding properties of modern human influenza viruses primarily isolated in Vero and MDCK cells and chicken embryonated eggs,” Virology, 2003, vol. 313, pp. 473-480.
Nikitin, T. et al. “Epidemiological Studies of A/Hong Kong/68 Virus Infection in Dogs”, Bull. World Health Organ., 1972, pp. 471-479, vol. 47.
Nobusawa, E. et al. “Comparison of Complete Amino Acid Sequences and Receptor-Binding Properties Among 13 Serotypes of Hemagglutinins of Influenza A Viruses”, Virology, 1991, pp. 475-485, vol. 182, No. 2.
Ohwada, K. et al. “Experimental Infections of Dogs with Type C Influenza Virus” Microbiol. Immunol., 1986, pp. 451-460, vol. 30, No. 5.
Olsen, C.W. et al. “Immunogenicity and efficacy of baculovirus-expressed and DNA-based equine influenza virus hemagglutinin vaccines in mice” Vaccine, 1997, 15(10):1149-1156.
Oxburgh, L. et al. “Evolution of H3N8 Equine Influenza Virus from 1963 to 1991”, Virus Res. Nov. 1994, pp. 153-165, vol. 34(2). Abstract Only.
Patriarca, P. A. et al. “Lack of Significant Person-to-Person Spread of Swine Influenza-like Virus Following Fatal Infection in an Immunocompromised Child”, Am. J. Epidemiol., 1984, pp. 152-158, vol. 119, No. 2.
Payungporn, S. et al. “Influenza A Virus (H3N8) in Dogs with Respiratory Disease, Florida” Emerging Infectious Diseases, Jun. 2008, pp. 902-908, vol. 14, No. 6.
Peek, S.F., “Acute respiratory distress syndrome and fatal interstitial pneumonia associated with equine uinfluenza in a neonatal foal” Journal of Veterinary Internal Medicine, Jan. 2004, pp. 132-134, vol. 18, No. 1.
Peiris, J. S. et al. “Re-Emergence of Fatal Human Influenza A Subtype H5N1 Disease”, Lancet, 2004, pp. 617-619, vol. 363.
Peiris, M. et al. “Human Infection with Influenza H9N2”, Lancet, 1999, pp. 916-917, vol. 354.
Posnett, D. N. et al. “A Novel Method for Producing Anti-Peptide Antibodies”, J. Biol. Chem., 1988, pp. 1719-1725, vol. 263, No. 4.
Proceedings One Hundred and Eighth Annual Meeting of the United States Animal Health Association, Oct. 2004, pp. 1, 2, 247, and 296. www.usaha.org/meetings/2004/2004_USAHA_Proceedings.pdf
Putnam, B. “Two Illnesses Seen in Death of Dogs”, St. Petersburg Times, Feb. 10, 1999.
Ramanujan, K., “Contagious equine flu virus infecting dogs across U.S. is isolated by Cornell researchers.” Posted by Cornell University News Service, Sep. 27, 2005 www.news.cornell.edu/stories/Sept05?Dog_Flu.kr.html.
Reid, A. H. et al. “Evidence of an Absence: The Genetic Origins of the 1918 Pandemic Influenza Virus”, Nat. Rev. Microbiol., 2004, pp. 909-914, vol. 2.
Rowe, T. et al. “Detection of Antibody to Avian Influenza A (H5N1) Virus in Human Serum by Using a Combination of Serologic Assays”, J. Clin. Microbiol., 1999, pp. 937-943, vol. 37, No. 4.
Saiki, R. “Enzymatic Amplification of Beta-Globin Genomic Sequences and Restriction Site Analysis for Diagnosis of Sickle Cell Anemia”, Science, 1985, pp. 1350-1354, vol. 230.
Songserm, T., et al. “Avian Influenza H5N1 in Naturally Infected Domestic Cat.” Emerging Infectious Diseases, Apr. 2006, pp. 681-683, vol. 12, No. 4.
Subbarao, K. et al. “Characterization of an Avian Influenza A (H5N1) Virus Isolated from a Child with a Fatal Respiratory Illness”, Science, 1998, pp. 393-396, vol. 279.
Suzuki, Y. et al. “Sialic Acid Species as a Determinant of the Host Range of Influenza A Viruses”, J. Virol., 2000, pp. 11825-11831, vol. 74, No. 24.
Tam, J. P. et al. “Synthetic Peptide Vaccine Design: Synthesis and Properties of a High Density Multiple Antigenic Peptide System”, PNAS USA, 1988, pp. 5409-5413, vol. 85, No. 15.
Top, F. H. et al. “Swine Influenza A at Fort Dix, New Jersey (Jan.-Feb. 1976). IV. Summary and Speculation”, J. Infect. Dis., 1977, pp. S376-S380, vol. 136, Supp.
Troyer, H.L., “Canine Influenza Virus (‘Dog Flu’): A Summary of Clinical Cases at the Oradell Animal Hospitald, Paramus, New Jersey” Oct. 2005.
University of Florida News, UF Researchers: Equine Influenza Virus Likely Involved in Recent Respiratory Disease Outbreak in Racing Greyhounds, 2004, accessed online at http://news.ufl.edu/2004/04/22/racedogflu/ on Nov. 4, 2009.
Van Maanen, C. and A. Cullinane “Equine influenza virus infections: An update” Veterinary Quarterly, 2002, 24(2):79-94.
Velazco JG et al., “Surveillance of influenza in Houston, Texas, USA: gradual transition from A/Victoria/75 (H3N2) to A/Texas/77 (H3N2) predominance and antigenic characterization of “intermediate” strains” Bulletin of the World Health Organization, 1983, 61(2):345-352.
Vines, A. et al. “The Role of Influenza A Virus Hemagglutinin Residues 226 and 228 in Receptor Specificity and Host Range Restriction”, J. Virol., 1998, pp. 7626-7631, vol. 72, No. 9.
Von Grotthuss, M., et al. “Influenza Mutation from Equine to Canine.” Science, Mar. 3, 2006, pp. 124, vol. 311.
Wagener, J. S. “Role of Canine Parainfluenza Virus and Bordetella bronchiseptica in Kennel Cough”, Am. J. Vet. Res., 1984, pp. 1862-1866, vol. 45, No. 9.
Wagner, R. et al. “N-Glycans Attached to the Stem Domain of Haemagglutinin Efficiently Regulate Influenza A Virus Replication”, J. Gen. Virol., 2002, pp. 601-609, vol. 83.
Webby, R. et al. “Molecular Constraints to Interspecies Transmission of Viral Pathogens”, Nat. Med., 2004, pp. S77-S81, vol. 10, No. 12.
Webster, R. G. “Influenza: An Emerging Disease”, Emerg. Infect. Dis., 1998, pp. 436-441, vol. 4, No. 3.
Webster, R. G. et al. “Evolution and Ecology of Influenza A Viruses”, Microbiol. Rev., 1992, pp. 152-179, vol. 56, No. 1.
Weis, W. et al. “Structure of the Influenza Virus Haemagglutinin Complexed with Its Receptor, Sialic Acid”, Nature, 1988, pp. 426-431, vol. 333.
Womble, D. D. “GCG: The Wisconsin Package of Sequence Analysis Programs”, Methods Mol. Biol., 2000, pp. 3-22, vol. 132.
Wood et al., “Studies with Inactivated Equine Influenza Vaccine: 1. Serologioal Responses of Ponies to Graded Doses of Vaccine,” The Journal Hygiene, 1983, vol. 90, No. 3, pp. 371-384.
Wood, J.M. et al., “The standardization of inactivated equine influenza vaccines by single-radial immunodiffusion” Journal of Biological Standardization, Apr. 1983, pp. 133-136, vol. 11, No. 2.
WSAVA News, “Equine influenza virus jumps to dogs in Florida” Sep. 14, 2004, http://www.vetscite.org/publish/items/001864/index.html.
Xu, D. et al. “Systemic Induction ofa Potato Pin2 Promoter by Wounding, Methyl Jasmonate, and Abscisic Acid in Transgenic Rice Plants”, Plant Molecular Biology, 1993, pp. 573-588, vol. 22.
Yang, T-T. et al. “Optomized Codon Usage and Chromophore Mutations Provide Enhanced Sensitivity with the Green Fluorescent Protein”, Nucleic Acid Research, 1996, pp. 4592-4593, vol. 24, No. 22.
Yoon, K.-Y. et al. “Influenza Virus Infection in Racing Greyhounds”, Emerg. Infect. Dis., 2005, pp. 1974-1975, vol. 11, No. 12.
“The Detectives” Cornell Veterinary Medicine Newsletter, published by the College of Veterinary Medicine at Cornell University, Fall 2004.
GenBank Accession No. DQ124157 “Influenza A virus (A/canine/Florida/242/2003(H3N8)) hemagglutinin gene” Jan. 25, 2006.
GenBank Accession No. DQ124190 “Influenza A virus (A/canine/FIorida/43/2004(H3N8)) hemagglutinin gene” Jan. 25, 2006.
Yamamoto, T. “Of the bird flu epidemic situation (commentary)” Influenza, Apr. 2005, vol. 6, No. 2, pp. 147-152.
Daly, J.M., “Antigenic and genetic evolution of equine H3N8 influenza A viruses,” J. General Virology, 1996, vol. 77, pp. 661-671.
Daly, J.M. et al., “Current perspectives on control of equine influenza,” Vet. Res., 2004, vol. 35, pp. 411-423.
DQ222914, Influenza A virus (A/equine/Wisconsin/1/03(H3N8)) neuraminidase (NA) gene, complete eds., Oct. 29, 2005.
ABB17174, neuraminidase [Influenza A virus (A/equine/Wsconsin/1/03(H3N8))], Oct. 29, 2005.
GenBank Accession No. DQ124160, Influenza A virus (A/canine/Florida/242/2003(H3N8)) M1 and M2 (MP) gene, complete eds, Sep. 28, 2005.
GenBank Accession No. ABA42435, M1 [Influenza A virus (A/canine/Florida/242/2003(H3N8))], Sep. 28, 2005.
Lytle, C.D. and Sagripanti, J-L. “Predicted inactivation of viruses of relevance to biodefense by solar radiation” J. Virology, 2005, 79(22):14244-14252.
Chang, C.P. et al. “Influenza virus isolations from dogs during a human epidemic in Taiwan” Int. J. Zoon., 1976, vol. 3, pp. 61-64.
Isachenko, V.A. et al. “Antigenic characteristics of influenza viruses isolated from domestic animals and birds in the USSR” Inst. Virusol, AMN SSSR, 1973, vol. 18, No. 6, pp. 700-705, Abstract.
Minke, J.M. et al. “Equine viral vaccines: the past, present and future” Veterinary Research, BioMed Central, 2004, vol. 35, Issue 4, pp. 425-443.
GenBank Accession No. AAX23574.1 “PA polymerase [Influenza A virus (A/equine/Kentucky/5/2002(H3N8))]” Mar. 12, 2005.
GenBank Accession No. AAZ23561 “PB2, partial [Influenza A virus (A/canine/Florida/242/2003(H3N8))]” Jul. 26, 2016.
GenBank Accession No. AAZ23572 “PB1 [Influenza A virus (A/canine/Florida/242/2003(H3N8))]” Jan. 25, 2006.
GenBank Accession No. AAZ23569 “PA [Influenza A virus (A/canine/Florida/242/2003(H3N8))]” Jan. 25, 2006.
GenBank Accession No. AAZ23580 “NP [Influenza A virus (A/canine/Florida/242/2003(H3N8))]” Jan. 25, 2006.
GenBank Accession No. ABA39852 “neuraminidase, partial [Influenza A virus (A/canine/Florida/242/2003(H3N8))]” Jul. 26, 2016.
Steinhauer, D. A. et al. “Role of Hemagglutinin Cleavage for the Pathogenicity of Influenza Virus” Virology, 1999, pp. 1-20, vol. 258.
Related Publications (1)
Number Date Country
20220105171 A1 Apr 2022 US
Provisional Applications (6)
Number Date Country
60779080 Mar 2006 US
60761451 Jan 2006 US
60759162 Jan 2006 US
60754881 Dec 2005 US
60728449 Oct 2005 US
60673443 Apr 2005 US
Divisions (4)
Number Date Country
Parent 15888715 Feb 2018 US
Child 16382429 US
Parent 14507332 Oct 2014 US
Child 15888715 US
Parent 13066651 Apr 2011 US
Child 14507332 US
Parent 11584818 Oct 2006 US
Child 13066651 US
Continuations (1)
Number Date Country
Parent 16382429 Apr 2019 US
Child 17471370 US
Continuation in Parts (1)
Number Date Country
Parent 11409416 Apr 2006 US
Child 11584818 US