Title of Invention

CHIMERIC INFECTIOUS DNA CLONES, CHIMERIC PORCINE CIRCOVIRUSES AND USES THEREOF.

Abstract The present invention relates to infectious DNA clones, infectious chimeric DNA clones of porcine circovirus (PCV), vaccines and means of protecting pigs against viral infection or postweaning multisystemic wasting syndrome (PMWS) caused by PCV2). The new chimeric infectious DNA clone and its derived, a virulent chimeric virus are constructed from the nonpathogenic PCV1 in which the immunogenic ORF gene of the pathogenic PCV2 replaces a gene of the nonpathogenic PCV1, preferably in the same position. The chimeric virus advantageously retains the nonpathogenic phenotype of PCV1 but elicits specific immune responses against the pathogenic PCV2. The invention further embraces the immunogenic polypeptide expression products (FIG. - 6)
Full Text CHIMERIC INFECTIOUS DNA CLONES, CHIMERIC
PORCINE CIRCOVIRUSES AND USES THEREOF
CROSS-REFERENCE TO RELATED U.S. APPLICATIONS
This nonprovisional application claims the benefit under 35 U.S.C. § 119(e) of U.S.
Provisional Application No. 60/424,840, filed on November 8, 2002, which claims the
benefit under 35 U.S.C. § 119(e) of U.S. Provisional Application No. 60/340,775, filed on
December 12, 2001. The two prior applications are incorporated herein by reference in their
entirety.
STATEMENT REGARDING FEDERALLY SPONSORED
RESEARCH OR DEVELOPMENT
Not Applicable
REFERENCE TO A "Sequence Listing"
The material on a single compact disc containing a Sequence Listing file provided in
this application is incorporated by reference. The date of creation is January 27, 2003 and
the size is approximately 9.5 kb.
BACKGROUND OF THE INVENTION
Field of the Invention
The present invention concerns infectious porcine circovirus type-1 (PCV1) and type-
2 (PCV2) DNA clones, chimeric PCV1-2 infectious DNA clones and live chimeric viruses
derived from the chimeric DNA clones, useful as vaccines.

Description of the Related Art
All patents and publications cited in this specification are hereby incorporated by
reference in their entirety.
Porcine circovirus (PCV) was originally isolated as a cell culture contaminant of a
porcine kidney cell line PK-15 (I. Tischer et al., "A very small porcine virus with circular
single-stranded DNA," Nature 295:64-66 (1982); I. Tischer et al., "Characterization of
papovavirus and picornavirus-like particles in permanent pig kidney cell lines," Zentralbl.
Bakteriol. Hyg. Otg. A. 226(2):153-167 (1974)). PCV is a small icosahedral non-enveloped
virus that contains a single stranded circular DNA genome of about 1.76 kb. PCV is
classified in the family of Circoviridae, which consists of three other animal circoviruses
(chicken anemia virus (CAV), psittacine beak and feather disease virus (PBFDV) and the
recently discovered columbid circovirus (CoCV) from pigeons) and three plant circoviruses
(banana bunchy top virus, coconut foliar decay virus and subterranean clover stunt virus) (M.
R. Bassami et al, "Psittacine beak and feather disease virus nucleotide sequence analysis and
its relationship to porcine circovirus, plant circoviruses, and chicken anemia virus," Virology
249:453-459 (1998); J. Mankertz et al., "Transcription analysis of porcine circovirus (PCV),"
Virus Genes 16:267-276 (1998); A. Mankertz et at., "Cloning and sequencing of columbid
circovirus (CoCV), a new circovirus from pigeons," Arch. Virol. 145:2469-2479 (2000); B.
M. Meehan et al., "Sequence of porcine circovirus DNA: affinities with plant circoviruses,"
J. Gen. Virol. 78:221-227 (1997); B. M. Meehan et al, "Characterization of novel circovirus
DNAs associated with wasting syndromes in pigs," J. Gen. Virol. 79:2171-2179 (1998); D.
Todd et al, "Comparison of three animal viruses with circular single-stranded DNA
genomes," Arch. Virol. 117:129-135 (1991)). Members of the three previously recognized
animal circoviruses (PCV, CAV, and PBFDV) do not share nucleotide sequence homology
or antigenic determinants with each other (M. R. Bassami et al., 1998, supra; D. Todd et al.,
1991, supra). The genome of the newly identified CoCV shared about 40% nucleotide
sequence identity with that of PCV (A. Mankertz et al, "Cloning and sequencing of
columbid circovirus (CoCV), a new circovirus from pigeons," Arch. Virol. 145:2469-2479
(2000)). Recently, a novel human circovirus with a circular genome, designated as
transfusion transmitted virus or TT virus (TTV), was identified from individuals associated
with post-transfusion hepatitis (H. Miyata et al, "Identification of a novel GC-rich 113-

nucleotide region to complete the circular, single-stranded DNA genome of TT virus, the
first human circovirus," J. Virol. 73:3582-3586 (1999); T. Nishizawa et al., "A novel DNA
virus (TTV) associated with elevated transaminase levels in posttransfusion hepatitis of
unknown etiology," Biochem. Biophys. Res. Commun. 241:92-97 (1997)). Additionally, a
human TTV-like mini virus (TLMV) was identified from normal blood donors (P. Biagini et
al., "Genetic analysis of full-length genomes and subgenomic sequences of TT virus-like
mini virus human isolates," J. Gen. Virol. 82: 379-383 (2001); K. Takahashi et al.,
"Identification of a new human DNA virus (TTV-like mini virus, TLMV) intermediately
related to TT virus and chicken anemia virus," Arch. Virol. 145:979-93 (2000)) and a third
novel human circovirus, known as SEN virus (SENV), was also discovered from humans
with post-transfusion hepatitis (T. Umemura et al., "SEN virus infection and its relationship
to transfusion-associated hepatitis," Hepathology 33:1303-1311 (2001)). The genomic
organization of both human TTV and TLMV is similar to that of the CAV (P. Biagini et al,
2001, supra; H. Miyata et al., 1999, supra; K. Takahashi et al., 2000, supra). Although
antibodies to PCV were found in various animal species including humans, mice, cattle and
pigs (G. M. Allan et al, "Production, preliminary characterization and applications of
monoclonal antibodies to porcine circovirus," Vet. Immunol. Immunopathol. 43:357-371
(1994); G. C. Dulac and A. Afshar, "Porcine circovirus antigens in PK-15 cell line (ATCC
CCL-33) and evidence of antibodies to circovirus in Canadian pigs," Can. J. Vet. Res.
53:431-433 (1989); S. Edwards and J. J. Sands, "Evidence of circovirus infection in British
pigs," Vet. Rec. 134:680-1 (1994); J. C. Harding and E.G. Clark, "Recognizing and
diagnosing postweaning multisystemic wasting syndrome (PMWS)," Swine Health and
Production 5:201-203 (1997); R. K. Hines and P. D. Lukert, "Porcine circovirus: a
serological survey of swine in the United States," Swine Health and Production 3:71-73
(1995); G. P. Nayar et al, "Evidence for circovirus in cattle with respiratory disease and
from aborted bovine fetuses," Can. Vet. J. 40:277-278 (1999); I. Tischer et al., "Distribution
of antibodies to porcine circovirus in swine populations of different breeding farms," Arch.
Virol. 140:737-743 (1995); I. Tischer et al, "Presence of antibodies reacting with porcine
circovirus in sera of humans, mice, and cattle," Arch. Virol. 140:1427-1439 (1995)), little is
known regarding the pathogenesis of PCV in these animal species. Experimental infection of
pigs with the PK-15 cells-derived PCV did not produce clinical disease and thus, this virus is

not considered to be pathogenic to pigs (G. M. Allan et al., "Pathogenesis of porcine
circovirus; experimental infections of colostrum deprived piglets and examination of pig
foetal material," Vet. Microbiol. 44:49-64 (1995); I. Tischer et al., "Studies on epidemiology
and pathogenicity of porcine circovirus," Arch. Virol. 91:271-276 (1986)). The
nonpathogenic PCV derived from the contaminated PK-15 cell line was designated as
porcine circovirus type 1 or PCV1.
Postweaning multisystemic wasting syndrome (PMWS), first described in 1991 (J. C.
Harding and E.G. Clark, 1997, supra), is a complex disease of weaning piglets that is
becoming increasingly more widespread. With the threat of a potential serious economic
impact upon the swine industry, it has become urgent to develop a vaccine against PCV2, the
primary causative agent of PMWS. PMWS mainly affects pigs between 5-18 weeks of age.
Clinical PMWS signs include progressive weight loss, dyspnea, tachypnea, anemia, diarrhea,
and jaundice. Mortality rate may vary from 1% to 2%, and up to 40% in some complicated
cases in the U.K. (M. Muirhead, "Sources of information on PMWS/PDNS," Vet. Rec.
150:456 (2002)). Microscopic lesions characteristic of PMWS include granulomatous
interstitial pneumonia, lymphadenopathy, hepatitis, and nephritis (G. M. Allan and J. A.
Ellis, "Porcine circoviruses: a review," J. Vet. Diagn. Invest. 12:3-14 (2000); J. C. Harding
and E.G. Clark, 1997, supra). PMWS has now been recognized in pigs in Canada, the
United States (G. M. Allan et al., "Novel porcine circoviruses from pigs with wasting disease
syndromes," Vet. Rec. 142:467-468 (1998); G. M. Allan et al., "Isolation of porcine
circovirus-like viruses from pigs with a wasting disease in the USA and Europe," J. Vet.
Diagn. Invest. 10:3-10 (1998); G. M. Allan and J. A. Ellis, 2000, supra; J. Ellis et al,
"Isolation of circovirus from lesions of pigs with postweaning multisystemic wasting
syndrome," Can. Vet. J. 39:44-51 (1998); A. L. Hamel et al., "Nucleotide sequence of
porcine circovirus associated with postweaning multisystemic wasting syndrome in pigs," J.
Virol. 72:5262-5267 (1998); M. Kiupel et al, "Circovirus-like viral associated disease in
weaned pigs in Indiana," Vet. Pathol. 35:303-307 (1998); R. Larochelle etal, "Identification
and incidence of porcine circovirus in routine field cases in Quebec as determined by PCR,"
Vet. Rec. 145:140-142 (1999); B. M. Meehan et al, 1998, supra; I. Morozov et al,
"Detection of a novel strain of porcine circovirus in pigs with postweaning multisystemic
wasting syndrome," J. Clin. Microbiol. 36:2535-2541 (1998)), most European countries (G.

M. Allan et al., "Isolation of porcine circovirus-like viruses from pigs with a wasting disease
in the USA and Europe," J. Vet. Diagn. Invest. 10:3-10 (1998); G. M. Allan and J. A. Ellis,
2000, supra; S. Edwards and J. J. Sands, 1994, supra; S. Kennedy et al., "Porcine circovirus
infection in Northern Ireland," Vet. Rec. 142:495-496 (1998); A. Mankertz et al.,
"Characterization of PCV-2 isolates from Spain, Germany and France," Virus Res. 66:65-77
(2000); C. Rosell et al, "Identification of porcine circovirus in tissues of pigs with porcine
dermatitis and nephropathy syndrome. Vet. Rec. 146:40-43 (2000); P. Spillane et al,
"Porcine circovirus infection in the Republic of Ireland," Vet. Rec. 143:511-512 (1998); G. J.
Wellenberg et al., "Isolation and characterization of porcine circovirus type 2 from pigs
showing signs of post-weaning multisystemic wasting syndrome in the Netherlands," Vet.
Quart. 22:167-72 (2000)) and some countries in Asia (C. Choi et al, "Porcine postweaning
multisystemic wasting syndrome in Korean pig: detection of porcine circovirus 2 infection by
immunohistochemistry and polymerase chain reaction," J. Vet. Diagn. Invest. 12:151-153
(2000); A. Onuki et al, "Detection of porcine circovirus from lesions of a pig with wasting
disease in Japan," J. Vet. Med. Sci. 61:1119-1123 (1999)). PMWS potentially has a serious
economic impact on the swine industry worldwide.
The primary causative agent of PMWS is a pathogenic strain of PCV designated as
porcine circovirus type 2 or PCV2 (G. M. Allan et al, "Novel porcine circoviruses from pigs

with wasting disease syndromes," Vet. Rec. 142:467-468 (1998); G. M. Allan et al.,
"Isolation of porcine circovirus-like viruses from pigs with a wasting disease in the USA and
Europe," J. Vet. Diagn. Invest. 10:3-10 (1998); G. M. Allan et al., "Isolation and
characterisation of circoviruses from pigs with wasting syndromes in Spain, Denmark and
Northern Ireland," Vet. Microbiol. 66:115-23 (1999); G. M. Allan and J. A. Ellis, 2000,
supra; J. Ellis el al., 1998, supra; A. L. Hamel et al., 1998, supra; B. M. Meehan et al., 1998,
supra; I. Morozov et al., 1998, supra). The complete genomic sequence of the PMWS-
associated PCV2 has been determined (M. Fenaux et al, "Genetic characterization of type 2
porcine circovirus (PCV-2) from pigs with postweaning multisystemic wasting syndrome in
different geographic regions of North America and development of a differential PCR-
restriction fragment length polymorphism assay to detect and differentiate between infections
with PCV-1 and PCV-2," J. Clin. Microbiol. 38:2494-503 (2000); A. L. Hamel et al., 1998,

.supra; J. Mankertz et al., 1998, supra; B. M. Meehan et al., 1997, supra; B. M. Meehan et
al., 1998, supra; I. Morozov et al., 1998, supra).
PCV1 is ubiquitous in pigs but is not pathogenic to pigs. In contrast, the genetically
related PCV2 is pathogenic and causes PMWS in pigs. Sequence analyses reveals that the
PMWS-associated PCV2 shares only about 75% nucleotide sequence identity with the
nonpathogenic PCV1. The ORF2 gene of both the nonpathogenic PCV1 and the pathogenic
PCV2 encodes for the major immunogenic viral capsid protein (P. Nawagitgul et al,
"Modified indirect porcine circovirus (PCV) type 2-based and recombinant capsid protein
(ORF2)-based ELISA for the detection of antibodies to PCV," Immunol. Clin. Diagn. Lab
Immunol. 1:33-40 (2002); P. Nawagitgul et al., "Open reading frame 2 of porcine circovirus
type 2 encodes a major capsid protein," J. Gen. Virol. 81:2281-2287 (2000)).
Initial attempts to reproduce clinical PMWS in conventional pigs by PCV2
inoculation were unsuccessful (M. Balasch et al., "Experimental inoculation of conventional
pigs with tissue homogenates from pigs with post-weaning multisystemic wasting
syndrome," J. Comp. Pathol. 121:139-148 (1999); M. Fenaux et al., "Cloned Genomic DNA
of Type 2 Porcine Circovirus (PCV-2) Is Infectious When Injected Directly into the Liver
and Lymph Nodes of SPF Pigs: Characterization of Clinical Disease, Virus Distribution, and
Pathologic Lesions," J. Virol. 76:541-551 (2002)). Experimental reproduction of clinical
PMWS in gnotobiotic pigs and conventional pigs with tissue homogenates from pigs with
naturally occurring PMWS and with cell culture propagated PCV2 produced mixed results.
Clinical PMWS was reproduced in gnotobiotic (SPF) pigs and colostrum-deprived and
caesarian-derived pigs co-infected with PCV2 and porcine parvovirus (PPV) (G. M. Allan et
al., "Experimental reproduction of severe wasting disease by co-infection of pigs with
porcine circovirus and porcine parvovirus," J. Comp. Pathol. 121:1-11 (1999); S. Krakowka
et al., "Viral wasting syndrome of swine: experimental reproduction of postweaning
multisystemic wasting syndrome in gnotobiotic swine by coinfection with porcine circovirus
2 and porcine parvovirus," Vet. Pathol. 37:254-263 (2000)), and in PCV2-inoculated
gnotobiotic pigs when their immune system was activated by keyhole hemocyanin in
incomplete Freund"s adjuvant (S. Krakowka et al., "Activation of the immune system is the
pivotal event in the production of wasting disease in pigs infected with porcine circovirus-2
(PCV-2)," Vet. Pathol. 38:31-42 (2001)).

Clinical PMWS was also reproduced in cesarean derived/colostrum deprived pigs
(CD/CD) inoculated with PCV2 alone (P. A. Harms et al., "Experimental reproduction of
severe disease in CD/CD pigs concurrently infected with type 2 porcine circovirus and
porcine reproductive and respiratory syndrome virus," Vet. Pathol. 38:528-539 (2001)) and
in conventional pigs co-infected with PCV2 and either porcine parvovirus (PPV) or porcine
reproductive and respiratory syndrome virus (PRRSV) (A. Rivora et al., "Experimental
inoculation of conventional pigs with porcine reproductive and respiratory syndrome virus
and porcine circovirus 2," J. Virol. 76: 3232-3239 (2002)). In cases of the PRRSV/PCV2 co-
infection, the PMWS characteristic pathological signs such as lymphoid depletion,
granulomatous inflammation and necrotizing hepatitis are induced by PCV2 and not by
PRRSV (P. A. Harms et al., 2001, supra). However, clinical PMWS was not reproduced in
gnotobiotic pigs infected with PCV2 alone (G. M. Allan et al., "Experimental infection of
colostrums deprived piglets with porcine circovirus 2 (PCV2) and porcine reproductive and
respiratory syndrome virus (PRRSV) potentiates PCV2 replication," Arch. Virol. 145:2421-
2429 (2000); G. M. Allan et al., "A sequential study of experimental infection of pigs with
porcine circovirus and porcine parvovirus: immunostaining of cryostat sections and virus
isolation, J. Vet. Med. 47:81-94 (2000); G. M. Allan et al., "Experimental reproduction of
severe wasting disease by co-infection of pigs with porcine circovirus and porcine
parvovirus," J. Comp. Pathol. 121:1-11 (1999); M. Balasch et al., 1999, supra; J. Ellis et al.,
"Reproduction of lesions of postweaning multisystemic wasting syndrome in gnotobiotic
piglets," J. Vet. Diagn. Invest. 11:3-14 (1999); S. Kennedy et al., "Reproduction of lesions of
postweaning multisystemic wasting syndrome by infection of conventional pigs with porcine
circovirus type 2 alone or in combination with porcine parvovirus" J. Comp. Pathol. 122:9-24
(2000); S. Krakowka et al., 2001, supra; S. Krakowka et al., 2000, supra; R. M.
Pogranichnyy et al., "Characterization of immune response of young pigs to porcine
circovirus type 2 infection," Viral. Immunol. 13:143-153 (2000)). The virus inocula used in
these studies were either homogenates of tissues from pigs with naturally occurring PMWS,
or virus propagated in PK-15 cell cultures (G. M. Allan et al., "Experimental infection of
colostrums deprived piglets with porcine circovirus 2 (PCV2) and porcine reproductive and
respiratory syndrome virus (PRRSV) potentiates PCV2 replication," Arch. Virol. 145:2421-
2429 (2000); G. M. Allan et al., "A sequential study of experimental infection of pigs with

porcine circovirus and porcine parvovirus: immunostaining of cryostat sections and virus
isolation, J. Vet. Med. 47:81-94 (2000); G. M. Allan et al., "Experimental reproduction of
severe wasting disease by co-infection of pigs with porcine circovirus and porcine
parvovirus," J. Comp. Pathol. 121:1-11 (1999); M. Balasch et al., 1999, supra; J. Ellis et al.,
1999, supra, S. Kennedy et al., 2000, supra; S. Krakowka et al., 2001, supra; S. Krakowka et
al., 2000, supra; R. M. Pogranichnyy et al., 2000, supra). Since tissue homogenates may
contain other common swine agents such as PPV and porcine reproductive and respiratory
syndrome virus (PRRSV) (G. M. Allan et al., "Experimental infection of colostrums
deprived piglets with porcine circovirus 2 (PCV2) and porcine reproductive and respiratory
syndrome virus (PRRSV) potentiates PCV2 replication," Arch. Virol. 145:2421-2429 (2000);
G. M. Allan et al., "Experimental reproduction of severe wasting disease by co-infection of
pigs with porcine circovirus and porcine parvovirus," J. Comp. Pathol. 121:1-11 (1999); G.
M. Allan and J. A. Ellis, 2000, supra; J. A. Ellis et al., "Coinfection by porcine circoviruses
and porcine parvovirus in pigs with naturally acquired postweaning multisystemic wasting
syndrome," J. Vet. Diagn. Invest. 12:21-27 (2000); C. Rosell et al., 2000, supra), and since
the ATCC PK-15 cell line used for PCV2 propagation was persistently infected with PCVI
(G. C. Dulac and A. Afshar, 1989, supra), the clinical disease and pathological lesions
reproduced in those studies may not be solely attributable to PCV2 infection (G. M. Allan et
al., "Experimental infection of colostrums deprived piglets, with porcine circovirus 2 (PCV2)
and porcine reproductive and respiratory syndrome virus (PRRSV) potentiates PCV2
replication," Arch. Virol. 145:2421-2429 (2000); G. M. Allan et al., "A sequential study of
experimental infection of pigs with porcine circovirus and porcine parvovirus:
immunostaining of cryostat sections and virus isolation, J. Vet. Med. 47:81-94 (2000); G. M.
Allan et al, "Experimental reproduction of severe wasting disease by co-infection of pigs
with porcine circovirus and porcine parvovirus," J. Comp. Pathol. 121:1-11 (1999); G. M.
Allan and J. A. Ellis, 2000, supra; J. A. Ellis et al, 2000, supra).
Clinical PMWS has also been reproduced in PCV2-inoculated CDCD pigs when
vaccinated with Mycoplasma hyopneumoniae (G. M. Allan et al., "Immunostimulation, PCV-
2 and PMWS," Vet. Rec. 147:171-172 (2000)). Two recent field studies by G. M. Allan et
al., "Neonatal vaccination for Mycoplasma hyopneumoniae and postweaning multisystemic
wasting syndrome: a field trial," Pig J. 48:34-41 (2001), and S. C. Kyriakis et al., "The

effects of immuno-modulation on the clinical and pathological expression of postweaning
multisystemic wasting syndrome," J. Comp. Pathol. 126:38-46 (2002), tested the effect of
immuno-modulation by Mycoplasma hyqpneumoniae vaccine on the development of PMWS
in endemic herds, and showed a significant decrease in PMWS cases in unvaccinated groups
compared to the vaccinated animals. However, another recent study using conventional SPF
piglets under controlled laboratory conditions could not reproduce such an effect, suggesting
that vaccinations with M. hyopneumoniae may potentially influence the development of
clinical PMWS but. it is clearly a secondary role to a PCV2 infection. Based on these and
other studies, PCV2 is nevertheless considered to be the primary but not the exclusive
causative agent of PMWS.
The lack of an infectious virus stock of a biologically pure form of PCV2 has
impeded the understanding of PCV2 pathogenesis and the etiological role of PCV2 in
PMWS. Vaccinations against PPV and possibly PRRSV have not consistently been shown to
prevent the onset of PMWS in PCV2 infected pigs. Consequently, finding a safe yet potent
vaccine that specifically targets PMWS has been difficult. There is a definite art-recognized
need in the veterinary field to produce an efficacious, safe vaccine against PCV2 infections
and PMWS.
U.S. Patent No. 6,287,856 (Poet et al.) and WO 99/45956 concern nucleic acids from
psittacine beak and feather disease virus (BFDV), a circovirus that infects avian species, and
from porcine circovirus (PCV). The patent proposes vaccine compositions comprising naked
DNA or mRNA and discloses a nucleic acid vector for the transient expression of PCV in a
eukaryotic cell comprising a cis-acting transcription or translation regulatory sequence
derived from the human cytomegalovirus immediate or early gene enhancer or promoter
functionally linked to a nucleic acid of the sequence. However, since the PCV DNA is
derived solely from the PK-15 cell line, it is likely to comprise the nonpathogenic PCV1
discovered nearly 30 years ago by I. Tischer et al., 1974, supra, and, therefore, it is not likely
to be effective in eliciting an immune reaction to PCV2 or infections caused by PCV2.
Subunit vaccines of recombinant proteins made from vectors comprising open reading
frames are also suggested in the patent but the open reading frames from PCV are not well
characterized or distinguished from each other. Since the source of the PCV DNA is PK-15

cells, the proteins made from those vectors comprising the open reading frames of PCV1
would not possess reliable immunogenic properties, if any, against PCV2.
U.S. Patent No. 6,217,883 (Allan et al.) and French Patent No. 2,781,159B relate to
the isolation of five PCV strains from pulmonary or ganglionic samples taken from pigs
infected with PMWS in Canada, California and France (Brittany), and their use in
combination with at least one porcine parvovirus antigen in immunogenic compositions.
Proteins encoded by PCV2 open reading frames (ORF) consisting of ORF1 to ORF13 are
broadly described in the patent but there is no exemplification of any specific protein
exhibiting immunogenic properties. The patent further discloses vectors consisting of DNA
plasmids, linear DNA molecules and recombinant viruses that contain and express in vivo a
nucleic acid molecule encoding the PCV antigen. Several other references, for example, U.S.
Patent No. 6,391,314.Bl; U.S. Patent No. 6,368,601 Bl; French Patent No. 2,769,321;
French Patent No. 2,769,322; WO 01/96377 A2; WO 00/01409; WO 99/18214; WO
00/77216 A2; WO 01/16330 A2; WO 99/29871; etc., also describe the administration of
PCV1 or PCV2 polypeptides or the nucleic acids encoding the polypeptides of various
strains.
However, the nonpathogenic PCV1 will not be useful against PCV2 infections and
the pathogenic PCV2 strains described in the art, even if attenuated, are likely to be of
limited value due to the usual tendency of a live virus to revert to its virulent state. Therefore,
there is still a long-standing need in the art for a live, infectious, nonpathogenic antigen for
the inoculation of pigs against serious infection or PMWS caused by PCV2 that is efficacious
and remains safe in veterinary vaccines. These goals are met by the construction of the new
live chimeric porcine circovirus described herein, which is based upon the genomic backbone
of the nonpathogenic PCV1 isolated by I. Tischer et al. almost 30 years ago. The novel
chimeric porcine circovirus of the present invention is able to satisfy that long-standing need
by uniquely and advantageously retaining the nonpathogenic phenotype of PCV1 but
eliciting specific immune response against pathogenic PCV2.
BRIEF SUMMARY OF THE INVENTION
The present invention concerns infectious chimeric DNA clones of porcine circovirus
(PCV) and live chimeric viruses derived from the DNA clones that are useful as vaccines.

The new live chimeric, genetically avirulent viruses are made from the nonpathogenic PCV1
genornic structure in which an immunogenic gene of a pathogenic PCV2 strain replaces a
gene of the PCV1, typically in the same corresponding position. The invention encompasses
the biologically functional plasmids, viral vectors and the like that contain the new
recombinant nucleic acid molecules described herein, suitable host cells transfected by the
vectors comprising the DNA and the immunogenic polypeptide expression products. Also
included within the scope of the present invention is a novel method of protecting pigs against
viral infection or postweaning multisystemic wasting syndrome (PMWS) caused by PCV2
comprising administering to a pig in need of such protection an immunologically effective
amount of a vaccine comprising, for example, the cloned chimeric DNA in a plasmid, a
chimeric virus derived from the chimeric DNA clone, the polypeptide products expressed from
the DNA described herein, etc. The invention further provides new infectious PCV2 molecular
DNA and reciprocal chimeric DNA clones of PCV that find use as experimental models in
obtaining or characterizing the novel avirulent viral vaccines.
BRIEF DESCRIPTION OF THE ACCOMPANYING DRAWINGS
The background of the invention and its departure from the art will be further
described hereinbelow with reference to the accompanying drawings, wherein:
Figure 1 represents the construction of an infectious PCV2 molecular DNA clone.
The relative positions of the primer pair used to amplify the complete PCV2 genome are
indicated by the arrows (reverse primer PCVS AC2, forward primer PCVSAC2). The PCV2
genomic DNA amplified by PCR is digested with SacII restriction enzyme, and purified.
The purified and SacII-digested genomic DNA is ligated to form concatemers. Ligated
concatemers are separated by gel electrophoresis, the tandem genome dimer of PCV2 is
purified and cloned into pSK vector that is pre-digested with SacII enzyme to produce a
molecular PCV2 DNA clone.
Figures 2A and 2B illustrate that the cloned PCV2 plasmid DNA is infectious when
transfected in vitro in PK-15 cells. Figure 2A shows the detection of PCV2 antigen by
immunofluorescence assay (IFA) in PK-15 cells transfected with the cloned PCV2 plasmid
DNA. Intense immunolabeling of PCV2 antigen is visualized in the nucleus, and to a lesser
degree, cytoplasm of the transfected cells. Figure 2B shows mock-transfected PK-15 cells.

Figure 3A shows the lungs from a pig inoculated by intralymphoid route with PCV2
DNA at 21 DPI. The lungs are rubbery, failed to collapse, and are mottled tan-red.
Tracheobronchial lymph nodes are markedly enlarged and tan (arrows). Figure 3B represents
a microscopic section of a normal lung from a control pig (25X). Figure 3C represents a
microscopic section of the lung from the pig in Figure 3A. Note the peribronchiolar
lymphohistiocytic inflammation and mild necrotizing bronchiolitis (25X). Figure 3D
illustrates the immunohistochemical staining of the lung in Figure 3A; Note the PCV2
antigen in macrophages (arrows) and fibroblast-like cells (arrow heads) around airways
(64X).
Figure 4A shows a normal lymph node from a control pig. Note the well-defined
lymphoid follicles (arrows) (25X). Figure 4B represents a microscopic section of the
tracheobronchial lymph node from the pig in Figure 3A inoculated 21 days previously by
intralymphoid route with cloned PCV2 genomic DNA. Lymphoid follicles are poorly
defined, there is mild-to-moderate lymphoid depletion, and mild multifocal granulomatous
inflammation (25X). Figure 4C represents a microscopic section of the lymph node in Figure
4B in a larger magnification focusing on one follicle. Note the poorly defined follicle with
macrophages and giant cells (arrow) replacing follicular lymphocytes (64X). Figure 4D
illustrates the immunohistochemical detection of PCV2 antigen in the same lymph node as
Figure 4B in macrophages (arrows) and giant cells (small arrowheads), and dendritic-like
cells (large arrowheads) in the follicles (64X).
Figure 5 illustrates the construction of a chimeric PCV1-2 (PCV1/PCV2) DNA clone
with the nonpathogenic PCV1 genome carrying the immunogenic ORF2 capsid gene of the
pathogenic PCV2. The dimmerized DNA clone is used for in vitro transfection of PK-15
cells to produce live chimeric virus expressing ORF2 protein of PCV2, and in vivo animal
experiments to confirm activity.
Figure 6 represents the construction and organization of the infectious PCV1, PCV2,
chimeric PCV1-2 and reciprocal chimeric PCV2-1 molecular DNA clones. The PCV2 DNA
clone is constructed by ligating two full-length linear PCV2 genomes in tandem into the
Bluescript SK vector (pSK) by the general methods described previously (M. Fenaux et al.,
2002, supra). PCV1 DNA clone is constructed by ligating two full-length linear PCV1
genomes in tandem into pSK vector. Chimeric PCV1-2 DNA clone is constructed by

replacing the ORF2 capsid gene of PCVI with that of the PCV2 in the nonpathogenic PCV1
genomic backbone in pSK vector. Reciprocal chimeric PCV2-1 DNA clone is constructed
by replacing the 0RF2 capsid gene of the pathogenic PCV2 with that of the nonpathogenic
PCV1 in the pathogenic PCV2 genomic backbone in pSK vector. Both chimeric clones are
dimmers in pSK vector. The arrows represent the relative locations of the PCR primers for
the detection of PCV1, PCV2, PCV1-2 and PCV2-1 viremia in inoculated animals.
Figures 7A-7J demonstrate that the PCVI, PCV2, chimeric PCVI-2 and reciprocal
chimeric PCV2-1 DNA clones are infectious and express respective viral antigens when
transfected in vitro in PK-15 cells. The left panel (7A, 7C, 7E, 7G and 71) is stained with
monoclonal antibody against the PCV1 ORF2. The right panel (7B, 7D, 7F, 7H and 7J) is
stained with antibody against PCV2. Panels 7A and 7B are mock transfected PK-15 cells.
Panels 7C and 7D are PK-15 cells transfected with the PCV1 DNA clone. Panels 7E and 7F
are PK-15 cells transfected with the PCV2 DNA clone. Panels 7G and 7H. are PK-15 cells
transfected with the chimeric PCVI-2 DNA clone. Panels 71 and 7J are PK-15 cells
transfected with the reciprocal chimeric PCV2-1 DNA clone.
Figure 8 represents the full-length DNA sequence of the cloned PCV2 molecular
DNA (which corresponds to SEQ ID NO:1).
Figure 9 represents the full-length DNA sequence of the cloned chimeric PCV1-2
DNA (which corresponds to SEQ ID NO:2).
Figure 10 represents the DNA sequence of the immunogenic ORF2 capsid gene of the
cloned chimeric PCV1-2 DNA (which corresponds to SEQ ID NO:3).
Figure 11 represents the putative amino acid translation of the immunogenic ORF2
capsid gene of the chimeric PCV1-2 DNA (which corresponds to SEQ ID NO:4).
DETAILED DESCRIPTION OF THE INVENTION
In accordance with the present invention, there are provided infectious molecular and
chimeric nucleic acid molecules of porcine circovirus (PCV), live chimeric viruses produced
from the chimeric nucleic acid molecule and veterinary vaccines to protect pigs from viral
infection or postweaning multisystemic wasting syndrome (PMWS) caused by PCV2. The
invention further provides immunogenic polypeptide expression products that may be used as
vaccines.

The new avirulent, infectious chimeric DNA molecule of PCV (PCVl-2) comprises a
nucleic acid molecule encoding an infectious, nonpathogenic PCV1 that contains an
immunogenic open reading frame (ORF) gene of a pathogenic PCV2 in place of an ORF gene
in the PCV1 genome. The infectious chimeric PCVl-2 DNA clone preferably contains the
immunogenic capsid gene (ORF2) of the PCV2 DNA cloned in the genomic backbone of the
infectious, nonpathogenic PCV1 DNA clone. Generally, the capsid gene of the PCV2 DNA
replaces the ORF2 gene of the PCV1 DNA in the nonpathogenic PCV1 genomic structure
but it is contemplated that a variety of positional permutations may be constructed through
genetic engineering to obtain other avirulent or attenuated chimeric DNA clones. The reciprocal
chimeric infectious PCV2-1 DNA clone between PCV1 and PCV2 is disclosed as a control to
analyze the chimeric PCVl-2 clone of the invention and is constructed by replacing the
capsid gene of PCV2 with that of PCV1 in the backbone of the pathogenic PCV2 infectious
DNA clone. In addition to being an experimental model, the reciprocal chimeric PCV2-1
DNA clone may find use in making specially tailored vaccines.
The cloned genomic DNA of PCV2 described herein is shown to be in vitro and in
vivo infectious when transfected into PK-15 cells and given to pigs. The infectious PCV2
DNA clone produces pathological lesions characteristic of PMWS in pigs allowing for an
improved characterization of clinical disease and understanding of virus distribution in the
tissue cells. This new, readily reproducible pathogenic agent lends itself to the development
of a suitable vaccination program to prevent PMWS in pigs,
The novel chimeric PCVl-2 DNA clone is also infectious by both in vitro
transfection of PK-15 cells and in vivo administration to pigs. In transfected PK-15 cells, the
chimeric PCVl-2 DNA clone expresses the PCV2 capsid antigen (the immunogenic capsid
protein of PCV2) whereas the reciprocal chimeric PCV2-1 DNA clone expresses the PCV1
capsid antigen, which is demonstrated by immunofluorescence assay (IFA) using antibodies
specific to PCV1 or PCV2 capsid antigen. Seroconversion to PCV2-specific antibody is
detected in pigs inoculated with the infectious PCV2 clone as well as the chimeric PCVl-2
clone. Detecting the seroconversion to PCV2-specific antibody establishes that the chimeric
PCVl-2 DNA clone induces the PCV2-specific antibody in infected pigs and, consequently,
acts to protect inoculated pigs from infection with PCV2.

The below examples describe the evaluation of the immunogenicity and pathogenicity
of the chimeric DNA clones in inoculated pigs in more detail. Basically, seroconversions to
antibodies against PCV2 ORF2 antigen are detected in pigs inoculated with the PCV2 DNA
clone (Group 3) and the chimeric PCV1-2 DNA clone (Group 4). All of the pigs inoculated
with the PCV1 clone and the reciprocal chimeric PCV2-1 DNA clone (Groups 2 and 5,
respectively) seroconvert to the PCV1 antibody. The viruses recovered from selected pigs in
each group are partially sequenced and confirmed to be the authentic respective infectious
DNA clones used in the inoculation. Gross and microscopic lesions in various tissues of
animals inoculated with the PCV2 DNA clone are significantly more severe than those found
in pigs inoculated with PCVI, chimeric PCVI-2 and reciprocal chimeric PCV2-1 DNA
clones.
Surprisingly and advantageously, the chimeric PCV1-2 infectious DNA clone having
the immunogenic capsid gene (ORF2) of the pathogenic PCV2 cloned into the nonpathogenic
PCV1 genomic backbone induces a specific antibody response to the pathogenic PCV2
capsid antigen while it uniquely retains the nonpathogenic nature of PCVI in pigs. Animals
inoculated with the chimeric PCVI-2 infectious DNA clone develop a mild infection
resembling that of PCV1 inoculated animals while seroconverting to the antibody against the
ORF2 capsid protein of the pathogenic PCV2. The average length of viremia observed in
PCV1 and chimeric PCV1-2 inoculated animals is shorter, 0.625 weeks and 1 week
respectively, than that in pathogenic PCV2 inoculated animals which is about 2.12 weeks.
The lack of detectable chimeric PCVI-2 viremia in some inoculated animals does not affect
seroconversion to antibody against PCV2 ORF2 capsid protein in the PCV1-2 inoculated
pigs (Group 4). The results indicate that, even though the chimeric PCVI-2 viremia is short
or undetectable in some inoculated animals, the chimeric PCV1-2 virus is able to induce
antibody response against PCV2 ORF2 capsid protein. The special ability of the chimeric
PCV1-2 infectious DNA clone to induce the immune response specific to the pathogenic
PCV2 immunogenic ORF2 capsid protein yet remain nonpathogenic to pigs makes the
chimeric PCV1-2 clone particularly useful as a genetically engineered live-attenuated
vaccine and other types of vaccines.
The novel, purified and isolated nucleic acid molecules of this invention comprise the
full-length DNA sequence of the cloned chimeric PCV1-2 DNA set forth in SEQ ED NO:2,

shown in Figure 9 and deposited in the American Type Culture Collection under Patent
Deposit Designation PTA-3912; its complementary strand (i.e., reverse and opposite base
pairs) or the nucleotide sequences having at least 95% homology to the chimeric nucleotide
sequence (i.e., a significant active portion of the whole gene). Conventional methods that are
well known in the art can be used to make the complementary strands or the nucleotide
sequences possessing high homology, for instance, by the art-recognized standard or high
stringency hybridization techniques. The purified and isolated nucleic acid molecule
comprising the DNA sequence of the immunogenic capsicl gene of the cloned chimeric
PCV1-2 DNA is also set forth in SEQ ID NO:3 and Figure 10.
Suitable cells containing the chimeric nucleic acid molecule uniquely produce live,
infectious chimeric porcine circoviruses. The live, infectious chimeric virus is derived from the
chimeric DNA clone by transfecting PK-15 cells via in vitro and in vivo transfections as
illustrated herein. A preferred example of the cloned chimeric PCV1-2 DNA is the nucleotide
sequence set forth in SEQ ID NO:2 and Figure 9. The invention further envisions that the
chimeric virus is derived from the complementary strand or the nucleotide sequences having
a high homology, at least 95% homology, to the chimeric nucleotide sequence.
Also included within the scope of the present invention are biologically functional
plasmids, viral vectors and the like that contain the new recombinant nucleic acid molecules
described herein, suitable host cells transfected by the vectors comprising the chimeric and
molecular DNA clones and the immunogenic polypeptide expression products. A particularly
preferred immunogenic protein has the amino acid sequence set forth in SEQ ID NO:4 and
Figure 11. The biologically active variants thereof are further encompassed by the invention.
One of ordinary skill in the art would know how to modify, substitute, delete, etc., amino acid(s)
from the polypeptide sequence and produce biologically active variants that retain the same, or
substantially the same, activity as the parent sequence without undue effort.
To produce the immunogenic polypeptide products of this invention, the process may
include the following steps: growing, under suitable nutrient conditions, prokaryotic or
eucaryotic host cells transfected with the new recombinant nucleic acid molecules described
herein in a manner allowing expression of said polypeptide products, and isolating the desired
polypeptide products of the expression of said nucleic acid molecules by standard methods

known in the art. It is contemplated that the immunogenic proteins may be prepared by other
techniques such as, for example, biochemical synthesis and the like.
Vaccines of the chimeric viral and molecular DNA clones, and methods of using
them, are also included within the scope of the present invention. Inoculated pigs are
protected from serious viral infection and PMWS caused by PCV2. The novel method
protects pigs in need of protection against viral infection or PMWS by administering to the pig
an immunologically effective amount of a vaccine according to the invention, such as, for
example, a vaccine comprising an immunogenic amount of the chimeric PCV1-2 DNA, the
cloned chimeric virus, a plasmid or viral vector containing the chimeric DNA of PCVI-2, the
polypeptide expression products, the recombinant PCV2 DNA, etc. Other antigens such as
PRRSV, PPV, other infectious swine agents and immune stimulants may be given
concurrently to the pig to provide a broad spectrum of protection against viral infections.
The vaccines comprise, for example, the infectious chimeric PCV1-2 DNA, the
cloned PCV chimeric DNA genome in suitable plasmids or vectors such as, for example, the
pSK vector, an avirulent, live chimeric virus, an inactivated chimeric virus, etc. in
combination with a nontoxic, physiologically acceptable carrier and, optionally, one or more
adjuvants. The vaccine may also comprise the infectious PCV2 molecular DNA clone
described herein. The infectious chimeric PCV1-2 DNA, the plasmid DNA containing the
infectious chimeric viral genome and the live chimeric virus are preferred with the live
chimeric virus being most preferred. The avirulent, live viral vaccine of the present invention
provides an advantage over traditional viral vaccines that use either attenuated, live viruses
which run the risk of reverting back to the virulent state or killed cell culture propagated whole
virus which may not induce sufficient antibody immune response for protection against the viral
disease.
The adjuvant, which may be administered in conjunction with the vaccine of the
present invention, is a substance that increases the immunological response of the pig to the
vaccine. The adjuvant may be administered at the same time and at the same site as the
vaccine, or at a different time, for example, as a booster. Adjuvants also may advantageously
be administered to the pig in a manner or at a site different from the manner or site in which
the vaccine is administered. Suitable adjuvants include, but are not limited to, aluminum
hydroxide (alum), immunostimulating complexes (ISCOMS), non-ionic block polymers or

copolymers, cytokines (like EL-1, EL-2, EL-7, IFN-?, IFN-?, IFN-?, etc.), saponins,
monophosphoryl lipid A (MLA), muramyl dipeptides (MDP) and the like. Other suitable
adjuvants include, for example, aluminum potassium sulfate, heat-labile or heat-stable
enterotoxin isolated from Escherichia coli, cholera toxin or the B subunit thereof, diphtheria
toxin, tetanus toxin, pertussis toxin, Freund"s incomplete or complete adjuvant, etc. Toxin-
based adjuvants, such as diphtheria toxin, tetanus toxin and pertussis toxin may be
inactivated prior to use, for example, by treatment with formaldehyde.
The vaccines may further contain additional antigens to promote the immunological
activity of the infectious chimeric PCV DNA clones such as, for example, porcine
reproductive and respiratory syndrome virus (PRRSV), porcine parvovirus (PPV), other
infectious swine agents and immune stimulants.
The new vaccines of this invention are not restricted to any particular type or method
of preparation. The cloned viral vaccines include, but are not limited to, infectious DNA
vaccines (i.e., using plasmids, vectors or other conventional carriers to directly inject DNA
into pigs), live vaccines, modified live vaccines, inactivated vaccines, subunit vaccines,
attenuated vaccines, genetically engineered vaccines, etc. These vaccines are prepared by
standard methods known in the art.
The live viral vaccine is generally the most desirable vaccine in that all possible
immune responses are activated in the recipient of the vaccine, including systemic, local,
humoral and cell-mediated immune responses. A killed vaccine, on the other hand, can only
induce humoral immune response. Albeit the most desirable, however, live viral vaccines
have several disadvantages, such as the potential risk of contamination with live adventitious
viral agents or the risk that the virus may revert to virulence in the field. Remarkably, the
unique PCV1-2 chimeric DNA of the present invention overcomes those disadvantages.
Using only the immunogenic genes of the pathogenic PCV2, the chimeric DNA constructs a
live, replicating chimeric virus that is nonpathogenic yet elicits the complete, beneficial
immune responses of live viral vaccines against the pathogenic PCV2 virus. The live virus
vaccine based on the chimeric virus will have little chance, if any, for reversion to a
pathogenic phenotype. Thus, the new chimeric virus based on the structure of the
nonpathogenic PCV1 has a huge advantage over any recombinant PCV2 DNA virus, any

live, attenuated PCV2 vaccine or any other type of vaccine predicated solely on PCV2 for
immunity against the PCV2 infections.
Although the live viral vaccine is most preferred, other types of vaccines may be used
to inoculate pigs with the new chimeric virus and other antigens described herein. To prepare
inactivated virus vaccines, for instance, the virus propagation from the infectious DNA clone
is done by methods known in the art or described herein. Serial virus inactivation is then
optimized by protocols generally known to those of ordinary skill in the art.
Inactivated virus vaccines may be prepared by treating the chimeric virus derived
from the cloned PCV DNA with inactivating agents such as formalin or hydrophobic
solvents, acids, etc., by irradiation with ultraviolet light or X-rays, by heating, etc.
Inactivation is conducted in a manner understood in the art. For example, in chemical
inactivation, a suitable virus sample or serum sample containing the virus is treated for a
sufficient length of time with a sufficient amount or concentration of inactivating agent at a
sufficiently high (or low, depending on the inactivating agent) temperature or pH to
inactivate the virus. Inactivation by heating is conducted at a temperature and for a length of
time sufficient to inactivate the virus. Inactivation by irradiation is conducted using a
wavelength of light or other energy source for a length of time sufficient to inactivate the
virus. The virus is considered inactivated if it is unable to infect a cell susceptible to
infection.
The preparation of subunit vaccines typically differs from the preparation of a
modified live vaccine or an inactivated vaccine. Prior to preparation of a subunit vaccine, the
protective or antigenic components of the vaccine must be identified. Such protective or
antigenic components include certain amino acid segments or fragments of the viral capsid
proteins which raise a particularly strong protective or immunological response in pigs;
single or multiple viral capsid proteins themselves, oligomers thereof, and higher-order
associations of the viral capsid proteins which form virus substructures or identifiable parts
or units of such substructures; oligoglycosides, glycolipids or glycoproteins present on or
near the surface of the virus or in viral substructures such as the lipoproteins or lipid groups
associated with the virus, etc. Preferably, a capsid protein, such as the protein encoded by the
ORF2 gene, is employed as the antigenic component of the subunit vaccine. Other proteins
encoded by the infectious DNA clone may also be used. These immunogenic components are

readily identified by methods known in the art. Once identified, the protective or antigenic
portions of the virus (i.e., the "subunit") are subsequently purified and/or cloned by
procedures known in the art. The subunit vaccine provides an advantage over other vaccines
based on the live virus since the subunit, such as highly purified subunits of the virus, is less
toxic than the whole virus.
If the subunit vaccine is produced through recombinant genetic techniques,
expression of the cloned subunit such as the ORF2 (capsid) gene, for example, may be
optimized by methods known to those in the art (see, for example, Maniatis et al., "Molecular
Cloning: A Laboratory Manual," Cold Spring Harbor Laboratory, Cold Spring Harbor, MA.,
1989). If the subunit being employed represents an intact structural feature of the virus, such
as an entire capsid protein, the procedure for its isolation from the virus must then be
optimized. In either case, after optimization of the inactivation protocol, the subunit
purification protocol may be optimized prior to manufacture.
To prepare attenuated vaccines from pathogenic clones, the tissue culture adapted,
live, pathogenic PCV2 is first attenuated (rendered nonpathogenic or harmless) by methods
known in the art, typically made by serial passage through cell cultures. Attenuation of
pathogenic clones may also be made by gene deletions or viral-producing gene mutations.
Then, the attenuated PCV2 viruses may be used to construct additional chimeric PCV1-2
viruses that retain the nonpathogenic phenotype of PCVI but can vary in the strength of the
immunogenicity traits selected from the PCV2 genome through recombinant technology.
The most preferred vaccine employs the live chimeric virus DNA clone, in particular,
the clone containing the immunogenic genes of PCV2 cloned in the backbone of the
nonpathogenic PCV1. Advantageously, the live chimeric virus, which is naturally avirulent
when constructed through genetic engineering, does not require time-consuming attenuation
procedures. The virus uniquely serves as a live but nonpathogenic replicating virus that
produces immunogenic proteins against PCV2 during virus replication, which can then elicit
a full range of immune responses against the pathogenic PCV2.
As a further benefit, the preferred live chimeric virus of the present invention
provides a genetically stable vaccine that is easier to make, store and deliver than other types
of attenuated vaccines. Avirulent or attenuated vaccines based upon chimeric viruses are
generally considered as safe as, if not safer than, the traditionally modified live vaccines (J.

Arroyo et al., "Molecular basis for attenuation of neurovirulence of a yellow fever
Virus/Japanese encephalitis virus chimera vaccine (ChimeriVax-JE)," J. Virol. 75(2):934-942
(2001); F. Guirakhoo et al., "Recombinant chimeric yellow fever-dengue type 2 virus is
immunogenic and protective in nonhuman primates," J. Virol. 74(12):5477-5485 (2000); S.
Tang et al., "Toward a poliovirus-based simian immunodeficiency virus vaccine: correlation
between genetic stability and immunogenicity," J. Virol. 71(10):7841-7850 (1997)). For
example, the ChimeriVax-JE vaccine against Japanese encephalitis virus (JEV), which is a
genetically engineered derivative of the yellow fever virus vaccine YFV17D in which the
genes encoding the structural proteins prM and E of YFV17D are replaced with the
corresponding genes of the attenuated JEV SA14-14-2 strain, has been shown to be
genetically stable after prolonged passages both in vitro and in vivo (J. Arroyo et al., 2001,
supra). Another chimeric virus vaccine ChimeriVax-D2 against Dengue virus type 2, which
is an attenuated chimeric yellow fever (YF)-dengue type 2 (dengue-2) virus, has also been
found to be genetically stable; its sequences were reported to be unchanged after 18 passages
in Vero cells (F. Guirakhoo et al, 2000, supra).
Another preferred vaccine of the present invention utilizes suitable plasmids for
delivering the nonpathogenic chimeric DNA clone to pigs. In contrast to the traditional vaccine
that uses live or killed cell culture propagated whole virus, this invention provides for the direct
inoculation of pigs with the plasmid DNA containing the infectious chimeric viral genome.
Additional genetically engineered vaccines, which are desirable in the present
invention, are produced by techniques known in the art. Such techniques involve, but are not
limited to, further manipulation of recombinant DNA, modification of or substitutions to the
amino acid sequences of the recombinant proteins and the like.
Genetically engineered vaccines based on recombinant DNA technology are made,
for instance, by identifying alternative portions of the viral gene encoding proteins
responsible for inducing a stronger immune or protective response in pigs (e.g., proteins
derived from ORF3, ORF4, etc.). Such identified genes or immuno-dominant fragments can
be cloned into standard protein expression vectors, such as the baculovirus vector, and used
to infect appropriate host cells (see, for example, O"Reilly et al, "Baculovirus Expression
Vectors: A Lab Manual," Freeman & Co., 1992). The host cells are cultured, thus expressing

the desired vaccine proteins, which can be purified to the desired extent and formulated into a
suitable vaccine product.
If the clones retain any undesirable natural abilities of causing disease, it is also
possible to pinpoint the nucleotide sequences in the viral genome responsible for the
virulence, and genetically engineer the virus avirulent through, for example, site-directed
mutagenesis. Site-directed mutagenesis is able to add, delete or change one or more
nucleotides (see, for instance, Zoller et al., DNA 3:479-488, 1984). An oligonucleotide is
synthesized containing the desired mutation and annealed to a portion of single stranded viral
DNA. The hybrid molecule, which results from that procedure, is employed to transform
bacteria. Then double-stranded DNA, which is isolated containing the appropriate mutation,
is used to produce full-length DNA by ligation to a restriction fragment of the latter that is
subsequently transfected into a suitable cell culture. Ligation of the genome into the suitable
vector for transfer may be accomplished through any standard technique known to those of
ordinary skill in the art. Transfection of the vector into host cells for the production of viral
progeny may be done using any of the conventional methods such as calcium-phosphate or
DEAE-dextran mediated transfection, electroporation, protoplast fusion and other well-
known techniques (e.g., Sambrook et al., "Molecular Cloning: A Laboratory Manual," Cold
Spring Harbor Laboratory Press, 1989). The cloned virus then exhibits the desired mutation.
Alternatively, two oligonucleotides can be synthesized which contain the appropriate
mutation. These may be annealed to form double-stranded DNA that can be inserted in the
viral DNA to produce full-length DNA.
Genetically engineered proteins, useful in vaccines, for instance, may be expressed in
insect cells, yeast cells or mammalian cells. The genetically engineered proteins, which may
be purified or isolated by conventional methods, can be directly inoculated into pigs to confer
protection against viral infection or postweaning multisystemic wasting syndrome (PMWS)
caused by PCV2.
An insect cell line (like HI-FIVE) can be transformed with a transfer vector
containing nucleic acid molecules obtained from the virus or copied from the viral genome
which encodes one or more of the immuno-dominant proteins of the virus. The transfer
vector includes, for example, linearized baculovirus DNA and a plasmid containing the

desired polynucleotides. The host cell line may be co-transfected with the linearized
baculovirus DNA and a plasmid in order to make a recombinant baculovirus.
Alternatively, DNA from a pig suffering from PMWS, which encode one or more
capsid proteins, the infectious PCV2 molecular DNA clone or the cloned PCV chimeric
DNA genome can be inserted into live vectors, such as a poxvirus or an adenovirus and used
as a vaccine.
An immunologically effective amount of the vaccines of the present invention is
administered to a pig in need of protection against viral infection or PMWS. The
immunologically effective amount or the immunogenic amount that inoculates the pig can be
easily determined or readily titrated by routine testing. An. effective amount is one in which
a sufficient immunological response to the vaccine is attained to protect the pig exposed to
the virus which causes PMWS. Preferably, the pig is protected to an extent in which one to
all of the adverse physiological symptoms or effects of the viral disease are significantly
reduced, ameliorated or totally prevented.
The vaccine can be administered in a single dose or in repeated doses. Dosages may
range, for example, from 1 to 1,000 micrograms of the plasmid DNA containing the
infectious chimeric DNA genome (dependent upon the concentration of the immuno-active
component of the vaccine), but should not contain an amount of virus-based antigen
sufficient to result in an adverse reaction or physiological symptoms of viral infection.
Methods are known in the art for determining or titrating suitable dosages of active antigenic
agent based on the weight of the pig, concentration of the antigen and other typical factors.
Preferably, the infectious chimeric viral DNA clone is used as a vaccine, or a live infectious
chimeric virus can be generated in vitro and then the live chimeric virus is used as a vaccine.
In that case, 100 to 200 micrograms of cloned chimeric PCV DNA or about 10,000 50%
tissue culture infective dose (TCID50) of live chimeric virus can be given to a pig.
Desirably, the vaccine is administered to a pig not yet exposed to the PCV virus. The
vaccine containing the chimeric PCV1-2 infectious DNA clone or other antigenic forms
thereof can conveniently be administered intranasally, transdermally (i.e., applied on or at the
skin surface for systemic absorption), parenterally, etc. The parenteral route of administration
includes, but is not limited to, intramuscular, intravenous, intraperitoneal, intradermal (i.e.,
injected or otherwise placed under the skin) routes and the like. Since the intramuscular and

intradermal routes of inoculation have been successful in other studies using viral infectious
DNA clones (E. E. Sparger et al., "Infection of cats by injection with DNA of feline
immunodeficiency virus molecular clone," Virology 238:157-160 (1997); L. Willems et al.,
"In vivo transfection of bovine leukemia provirus into sheep," Virology 189:775-777 (1992)),
these routes are most preferred, in addition to the practical intranasal route of administration.
Although less convenient, it is also contemplated that the vaccine is given to the pig through
the intralymphoid route of inoculation. A unique, highly preferred method of administration
involves directly injecting the plasmid DNA containing PCV1-2 chimera into the pig
intramuscularly, intradermally, intralymphoidly, etc.
When administered as a liquid, the present vaccine may be prepared in the form of an
aqueous solution, syrup, an elixir, a tincture and the like. Such formulations are known in the
art and are typically prepared by dissolution of the antigen and other typical additives in the
appropriate carrier or solvent systems. Suitable carriers or solvents include, but are not
limited to, water, saline, ethanol, ethylene glycol, glycerol, etc. Typical additives are, for
example, certified dyes, flavors, sweeteners and antimicrobial preservatives such as
thimerosal (sodium ethylmercurithiosalicylate). Such solutions may be stabilized, for
example, by addition of partially hydrolyzed gelatin, sorbitol or cell culture medium, and
may be buffered by conventional methods using reagents known in the art, such as sodium
hydrogen phosphate, sodium dihydrogen phosphate, potassium hydrogen phosphate,
potassium dihydrogen phosphate, a mixture thereof, and the like.
Liquid formulations also may include suspensions and emulsions that contain
suspending or emulsifying agents in combination with other standard co-formulants. These
types of liquid formulations may be prepared by conventional methods. Suspensions, for
example, may be prepared using a colloid mill. Emulsions, for example, may be prepared
using a homogenizer.
Parenteral formulations, designed for injection into body fluid systems, require proper
isotonicity and pH buffering to the corresponding levels of porcine body fluids. Isotohicity
can be appropriately adjusted with sodium chloride and other salts as needed. Suitable
solvents, such as ethanol or propylene glycol, can be used to increase the solubility of the
ingredients in the formulation and the stability of the liquid preparation. Further additives
that can be employed in the present vaccine include, but are not limited to, dextrose,

conventional antioxidants and conventional chelating agents such as ethylenediamine
tetraacetic acid (EDTA). Parenteral dosage forms must also be sterilized prior to use.
Another embodiment of the present invention involves a new method of preparing an
infectious, nonpathogenic chimeric nucleic acid molecule of PCV1-2, which comprises
removing an open reading frame (ORF) gene of a nucleic acid molecule encoding an
infectious nonpathogenic PCVI, replacing the same position with an immunogenic ORF
gene of a nucleic acid molecule encoding an infectious pathogenic PCV2, and recovering the
chimeric nucleic acid molecule. The nucleic acid molecule is typically DNA. A preferred
method replaces the ORF2 gene of the nonpathogenic PCV1 DNA with the immunogenic
ORF2 capsid gene of the infectious pathogenic molecular DNA of PCV2 described herein. It
is contemplated that other ORF positions or immunogenic fragments thereof can be exchanged
between the PCV1 and PCV2 DNA to construct the attenuated infectious chimeric DNA clones
according to the methods described herein.
The recombinant nucleic acid molecule is then used to construct the live, infectious,
replicating chimeric virus of the present invention that advantageously retains the
nonpathogenic nature of PCV1 yet expresses the immunogenic ORP2 protein of the
pathogenic PCV2 and elicits a complete immune response against the pathogenic PCV2.
Desirably, the PCV1-2 DNA clone serves as a genetically engineered avirulent, live vaccine
against PCV2 infection and PMWS in pigs.
An infectious DNA clone of PCV2 is constructed, as described herein, so that a
biologically pure and homogeneous infectious virus stock can be generated for pathogenesis
studies and the development of nonpathogenic, chimeric vaccines. The course of clinical
disease, virus distribution and pathological lesions associated with PCV2 infection are more
definitively characterized by using this molecular DNA clone and a biologically pure and
homogeneous infectious PCV2 virus stock derived from the molecular DNA clone than have
been observed in the past, which lends itself to the development of the desired vaccine
products of the present invention.
The PCV2 molecular clone is generated by ligating two copies of the complete PCV2
genome in tandem into the pSK vector. In sharp contrast to the single copy genome
disclosed in the art, the infectious DNA PCV2 clone made by the methods described herein
contains two complete copies of the PCV2 genome ligated together in tandem repeat.

Ligating two copies of genome in tandem provides a similar circular genome that mimics the
usual circular genome of PCV2. The advantage of having two copies, of the genome in
tandem in the infectious DNA PCV2 clone is to be able to maximize replication when the
infectious DNA clone is transfected in vitro and in vivo. Thus, the clone of the invention
operates more efficiently and effectively than the prior single copy genome.
Infection of animals with the molecular viral clone is extremely useful to studying the
genetic determinants of viral replication and virulence in the host. Type-2 porcine circovirus
(PCV2) has been incriminated as the causative agent of postweaning multisystemic wasting
syndrome (PMWS). PMWS is a complex disease syndrome in swine and multiple factors
may be involved in the clinical presentation of PMWS. However, the difficulty in producing
a biologically pure form of PCV2 due to the presence of other common swine agents in the
tissue homogenates of diseased pigs has impeded a definitive characterization of the clinical
disease and pathological lesions solely attributable to PCV2 infection. This is the first time
an infectious molecular DNA clone of PCV2 has been constructed and used to characterize
the disease and pathological lesions associated with PCV2 infection by direct in vivo
transfection of pigs with the molecular clone.
The homogeneous PCV2 live virus stock derived from the molecular clone is shown
to be infectious in vitro when transfected into PK-15 cells. The cloned PCV2 genomic DNA
is also infectious when directly injected into the livers and superficial iliac lymph nodes of
specific-pathogen-free (SPF) pigs. Animals injected with the cloned PCV2 plasmid DNA
develop an infection and disease resembling that induced by intranasal inoculation with a
homogenous, infectious PCV2 live virus stock. Seroconversion to PCV2-specific antibody is
detected in the majority of pigs from the inoculated groups at 35 days postinoculation (DPI).
The onset and duration of viremia in pigs inoculated with the chimeric PCV1-2 DNA
clone are similar to those of the pigs inoculated with the nonpathogenic PCV1 DNA clone,
whereas viremia in pigs inoculated with the PCV2 clone appears earlier and lasted longer.
Beginning at 14 DPI and lasting about 2-4 weeks, viremia is detected in the majority of the
PCV2-inoculated animals. Similarly, the majority of inoculated pigs necropsied at 35 DPI
seroconverted to PCV2-antibodies. PCV2 antigen is detected in various tissues and organs in
inoculated pigs. Gross lesions are limited to the lungs and lymph nodes, and are
characterized by systematically enlarged tan colored lymph nodes, lungs that failed to

collapse and mild multifocal tan-colored foci of consolidation. Gross lesions affecting the
lymph nodes in both the nonpathogenic PCV1 and the chimeric PCV1-2 inoculated pigs are
mild and limited to only a few animals, whereas the pathogenic PCV2 inoculated pigs all
have moderate-to-severe swelling and discoloration of lymphoid tissues (Table 9, below).
Statistical analysis reveals that the scores of the gross lesions in the lymph nodes of the
chimeric PCV1-2 inoculated animals are similar to those in nonpathogenic PCV1 inoculated
pigs. At 21 DPI, PCV2 inoculated pigs have gross lesions that are statistically more severe
than those of the PCV1 and the chimeric PCV1-2 inoculated pigs. Histopathological lesions
and PCV2-specific antigen are detected in numerous tissues and organs including brain, lung,
heart, kidney, tonsil, lymph nodes, spleen, ileum and liver of the inoculated (infected) pigs.
The histopathological lesions in multiple tissues and organs similar to those of PMWS are
reproduced with the PCV2 molecular DNA clone as well as with the infectious virus
prepared in vitro from the molecular DNA clone. Microscopically, at both 21 and 49 DPIs,
the chimeric PCV1-2 inoculated animals have statistically less microscopic lesions than the
PCV2 inoculated animals. The microscopic lesion scores in lymph nodes of the chimeric
PCV1-2 inoculated pigs are similar to those of the nonpathogenic PCV1, the reciprocal
chimeric PCV2-1 and the uninoculated control animals. Moderate to severe microscopic
lesions are found in multiple tissues of pathogenic PCV2 inoculated animals including lung,
liver, lymphoid, spleen, brain, heart, kidney and tonsil tissue. However, in chimeric PCVI-2
inoculated animals, mild to moderate microscopic lesions are limited only to liver, lymph
nodes and kidney tissues (see Table 10, below).
There are no remarkable clinical signs of PMWS in the control or any of the
inoculated pigs. Although the characteristic clinical symptoms of PMWS are not observed
with the cloned PCV2 plasmid DNA (the infectious PCV2 DNA clone) or with a biologically
pure PCV2 infectious virus stock, PCV2 is clearly responsible for the PMWS-like
histopathological lesions reproduced in the below illustrative examples. It is generally
believed that PCV2 is the primary but not the sole pathogenic agent responsible for the onset
of clinical PMWS.
This invention more definitively characterizes the clinical course and pathological
lesions exclusively attributable to PCV2 infection. The present data in the below illustrative
examples indicate that the readily reproduced, cloned PCV2 genomic DNA is available to

replace infectious virus for the PCV2 pathogenesis and immunization studies. While PCV2 is
shown as essential for development of PMWS, other factors or agents such as PRRSV, PPV,
etc. may be required to induce the full spectrum of clinical signs and lesions associated with
advanced cases of PMWS. However, with the knowledge that PCV2 is a key factor, the
novel infectious, replicating viral clone of the present invention can be further modified or
genetically engineered to achieve the desired optimal immunogenic effect through methods
known to those of ordinary skill in immunology and molecular genetics.
The availability of the infectious DNA clone of PCV2 described herein makes it
feasible to develop the genetically engineered attenuated vaccine for preventing PCV2
infection and PMWS in pigs. It is known that PCV2 replicates in the lymph nodes, lungs and
liver during natural infection, and one of the major pathogenic effects is the impairment of
the immune system by degradation of the lymphoid structures (S. Krakowka et al., 2001,
supra; G. M. Allan and J. A. Ellis, 2000, supra; S. Kennedy et al., 2000, supra; G. J.
Wellenberg et al., 2000, supra; G. M. Allan et al., "Experimental reproduction of severe
wasting disease by co-infection of pigs with porcine circovirus and porcine parvovirus," J.
Comp. Pathol. 121:1-11 (1999); J. Ellis et al., "Reproduction of lesions of postweaning
multisystemic wasting syndrome in gnotobiotic piglets," J. Vet. Diagn. Invest. 11:3-14
(1999); J. C. Harding and E.G. Clark, 1997, supra). By using this novel infectious PCV2
molecular DNA clone, the clinical disease, pathological lesions and virus distribution
exclusively attributable to PCV2 infection are more definitively characterized.
The structural and functional relationships of the PCV genes are better understood
due to the availability of the PCV2, PCV1, chimeric PCVl-2, and reciprocal chimeric PCV2-
1 infectious DNA clones described herein. Will et al., "Cloned HBV DNA causes hepatitis
in chimpanzees," Nature 299:740-742 (1982), first demonstrated the feasibility of using a
cloned hepatitis B virus DNA to infect chimpanzees by direct in vivo injection. This
approach has since been used to study viral replication and pathogenesis of several other
viruses (T. W. Dubensky et al., "Direct transfection of viral and plasmid DNA into the liver
or spleen of mice," Proc. Natl. Acad. Sci. USA 81:7529-7533 (1984); R. Girones et al.,
"Complete nucleotide sequence of a molecular clone of woodchuck hepatitis virus that is
infectious in the natural host," Proc. Natl. Acad. Sci. USA 86:1846-1849 (1989); N. L. Letvin
et al, "Risks of handling HIV," Nature 349:573 (1991); C. Seeger et al, "The cloned

genome of ground squirrel hepatitis virus is infectious in the animal. Proc. Natl. Acad. Sci.
USA. 81:5849-5852 (1984); E. E. Sparger et al, "Infection of cats by injection with DNA of
feline immunodeficiency virus molecular clone," Virology 238:157-160 (1997); R. Sprengel
et al, "Homologous recombination between hepadnaviral genomes following in vivo DNA
transfection: implications for studies of viral infectivity," Virology 159:454-456 (1987); H.
Will et al, 1982, supra; L. Willems et al., "In vivo transfection of bovine leukemia provirus
into sheep," Virology 189:775-777 (1992)).
The construction of an infectious PCV2 molecular DNA clone, and the demonstration
of infection by direct injection of cloned PCV2 plasmid DNA into the liver and lymph nodes
of pigs in the context of the present invention are advantageous for PCV2 studies. This in
vivo transfection system will enhance the study of the structural and functional relationship
of PCV2 genes using recombinant plasmids constructed in vitro to test different regions or
genes of PCV2 for their roles in virus replication and pathogenesis in the host. The
replication and pathogenesis of PCV2 can be studied in vivo without having to produce
infectious virus stocks by propagating PCV2 in cell cultures. This is advantageous as serial
cell culture passages may select for viral variants. Another advantage of using cloned PCV2
genomic DNA, instead of live virus, for animal studies is its relative ease for quantitation of
the inoculation dose. The amount of the cloned PCV2 DNA used for animal inoculation can
be easily determined by a spectrophotometer, whereas the dose of live PCV2 virus requires
infectivity titration in cell cultures and confirmation of infection by IFA. Direct injection of
animals with cloned PCV2 plasmid DNA eliminates the problems associated with the
presence of other indigenous swine agents in tissue homogenate inocula in animal studies.
In the present invention, the immunogenic ORF2 capsid gene is switched between the
pathogenic PCV2 and the nonpathogenic PCV1 to produce the unique structure of the
chimeric PCV1-2 infectious DNA clone. Surprisingly and advantageously, the chimeric
PCV1-2 infectious clone replicated, expressed the immunogenic ORF2 capsid antigen in
vitro and in vivo, and induced a specific antibody response against PCV2 ORF2 but retained
the nonpathogenic nature of PCV1. The chimeric PCV1-2 infectious DNA clone has the
ability to induce a strong immune response against PCV2 while inducing only a limited
infection with mild pathologic lesions similar to that of the nonpathogenic PCV1. For
vaccine development, the relatively easy storage and stability of cloned DNA, and the

economy of large-scale recombinant PCV2 plasmid DNA and chimeric PCV1-2 DNA clone
production provides an attractive means of delivering a live,, infectious viral DNA vaccine or
genetically engineered, attenuated viral vaccines to pigs. Therefore, the chimeric PCV1-2
infectious DNA clone taught in this invention is a useful vaccine candidate against PCV2
infection and PMWS.
It should be appreciated that all scientific and technological terms used herein have
the same meaning as commonly understood by those of ordinary skill in the art. For purposes
of this invention, the term "infectious" means that the virus replicates in pigs, regardless of
whether or not the virus causes any diseases. "SPF" refers to Specific-pathogen-free pigs.
The "gnotobiotic" pigs intend germ-free pigs. The terms "PCV2 plasmid DNA," "PCV2
genomic DNA" and "PCV2 molecular DNA" are being used interchangeably to refer to the
same cloned nucleotide sequence.
The infectious PCV1/PCV2 chimeric DNA clone (strain designation "PCV1-2
chimera"), the infectious PCV2 molecular DNA clone (strain designation "PCV2 clone") and
the biologically pure and homogeneous PCV2 stock derived from an Iowa sample of PCV2
that had been isolated from a pig with severe PMWS and identified as isolate number 40895
(strain designation "PCV2 #40895") are deposited under the conditions mandated by 37
C.F.R. § 1.808 and maintained pursuant to the Budapest Treaty in the American Type
Culture Collection (ATCC), 10801 University Boulevard, Manassas, Virginia 20110-2209,
U.S.A. The DNA sequences described herein are contained within 6,490 bp plasmids cloned
into pBluescript SK(+) vector (pSK) (Stratagene Inc., La Jolla, CA) and transformed into
Escherichia coli DH5a competent cells. The plasmids containing the infectious chimeric
PCV1-2 DNA clone (identified as "chimeric porcine circovirus Type 1 (PCV1) and Type 2
(PCV2) infectious DNA clone") and the infectious PCV2 molecular DNA clone (identified
as "infectious DNA clone of Type 2 porcine circovirus (PCV2)") have been deposited in the
ATCC on December 7, 2001 and have been assigned ATCC Patent Deposit Designations
PTA-3912 and PTA-3913, respectively. It should be appreciated that other plasmids, which may
be readily constructed using site-directed mutagenesis and the techniques described herein, are
also encompassed within the scope of the present invention. The biologically pure and
homogeneous PCV2 sample of isolate number 40895 (identified as "Type 2 porcine circovirus
(PCV2)") has also been deposited in the ATCC on December 7, 2001 and has been assigned

ATCC Patent Deposit Designation PTA-3914. The genornic (nucleotide) sequence of the
PCV2 isolate number 40895 has been deposited with the Genbank database and has been
publicly available since July 23, 2000 under accession number AF264042.
The following examples demonstrate certain aspects of the present invention.
However, it is to be understood that these examples are for illustration only and do not
purport to be wholly definitive as to conditions and scope of this invention. It should be
appreciated that when typical reaction conditions (e.g., temperature, reaction times, etc.) have
been given, the conditions both above and below the specified ranges can also be used,
though generally less conveniently. The examples are conducted at room temperature (about
23°C to about 28°C) and at atmospheric pressure. All parts and percents referred to herein
are on a weight basis and all temperatures are expressed in degrees centigrade unless
otherwise specified.
A further understanding of the invention may be obtained from the non-limiting
examples that follow below.
EXAMPLE 1
Generation of a PK-15 Cell Line Free of PCV1 Contamination
The source of the PCV2 isolate was from a spleen tissue sample of a pig with
naturally occurring PMWS (PCV2 serial identification number 40895, referred to as "isolate
40895") (M. Fenaux et at., 2000, supra). Immunohistochemical staining (IHC) with PCV2-
specific antibody confirmed the presence of PCV2 antigen in the tissue. The spleen tissues
were stored at -80°C until use.
The PK-15 cell line purchased from the American Type Culture Collection (ATCC
accession number CCL-33) was persistently infected with PCV1 (G. C. Dulac and A. Afshar,
1989, supra). Since only a subpopulation of PK-15 cells was persistently infected (id.), a
PK-15 cell line that is free of PCV1 contamination by end-point dilution was generated.
Protocol proceeded as follows: PK-15 cells were grown in MEM with Earle"s salts and L-
glutamine (Life Technologies, Inc., Grand Island, NY) supplemented with 10% fetal bovine
serum (FBS) and 1X antibiotic (Life Technologies, Inc.). Confluent cell monolayers were
trypsinized, and the cells were then counted and serially diluted to an end point with one cell
per 0.2 ml. The end point dilution was plated in 96-well plates and allowed to grow into a

monolayer starting from a single cell. Cells from each well were tested for PCV1 DNA
using a PCR-RFLP assay capable of detecting and differentiating PCV1 and PCV2 (M.
Fenaux et al., 2000, supra). PK-15 cells from wells that were tested negative for PCV1 by
the PCR-RFLP assay were subsequently expanded. The PCV1 free PK-15 cell line was
subcultured five additional passages and was found negative for PCV1 DNA by PCR at each
passage.
Four cell lines that were negative for PCV1 contamination were produced by the end-
point dilution of the persistently infected PK-15 cells from ATCC. The cell lines remained
negative for PCV1 by PCR after the five additional passages. One of the cell lines was
subsequently expanded and was shown to be able to support PCV2 replication when the cells
were transfected with the PCV2 molecular DNA clone (Fig. 2) and infected with PCV2 virus.
The cloned cells were further used for the in vitro transfection of PCV2 molecular DNA
clone to generate a biologically pure PCV2 infectious virus stock for the animal inoculation
experiment.
EXAMPLE 2
Construction of the PCV2 Infectious DNA Clone
To construct a PCV2 molecular DNA clone, a pair of PCR primers was designed
according to the published sequence of the PCV2 isolate 40895 (M. Fenaux et al., 2000,
supra): forward primer F-PCVSAC2 (5"-GAACCGCGGGCTGGCTGAACTTTTGAAAGT-
3"), set forth in SEQ ID NO:5, and reverse primer R-PCVSAC2 (5"-
GCACCGCGGAAATTTCTGACAAACGTTACA-3"), set forth in SEQ ID NO:6. This pair
of primers amplifies the complete genome of PCV2 with an overlapping region containing
the unique SacII restriction enzyme site (Fig. 1). DNA was extracted using the QIAamp
DNA Minikit (Qiagen, Inc., Valencia, CA) from a spleen tissue sample of a pig with
naturally occurring PMWS (isolate 40895) (M. Fenaux et al., 2000, supra). The extracted
DNA was amplified by PCR with AmpliTaq Gold polymerase (Perkin-Elmer, Norwalk, CT).
The PCR reaction consisted of an initial enzyme activation step at 95°C for 9 min, followed
by 35 cycles of denaturation at 94°C for 1 min, annealing at 48°C for 1 min, extension at
72°C for 3 min, and a final extension at 72°C for 7 min. The PCR product of expected size

was separated by gel electrophoresis and purified with the glassmilk procedure with a
Geneclean Kit (Bio 101, Inc., La Jolla, CA).
To construct a molecular DNA clone containing a tandem dimer of PCV2 genome,
the PCR product containing the complete PCV2 genome was first ligated into the advanTAge
plasmid vector (Clontech, Palo Alto, CA). E. Coli DH5? competent cells were transformed.
The recombinant plasmids were verified by restriction enzyme digestion. The full length
PCV2 genomic DNA was excised from the advanTAge vector by digestion with SacII
restriction enzyme. The digested PCV2 genomic DNA was ligated with T4 DNA ligase at
37°C for only 10 min, which favors the production of tandem dimers. The tandem dimers
were subsequently cloned into pBluescript SK(+) vector (pSK) (Stratagene Inc., La Jolla,
CA) (Fig. 1). Recombinant plasmids containing tandem dimers of PCV2 genome (herein
referred to as PCV2 molecular DNA clone) were confirmed by PCR, restriction enzyme
digestion, and DNA sequencing. The DNA concentration of the recombinant plasmids was
determined spectrophotometricaly.
Specifically, the complete genome of the PCV2 (isolate 40895) was amplified by
PCR to construct the infectious PCV2 molecular DNA clone. Two copies of the complete
PCV2 genome were ligated in tandem into the pSK vector to produce the PCV2 molecular
DNA clone (Fig. 1). The infectivity of the PCV2 molecular DNA clone was determined by
in vitro transfection of the PK-15 cells. IFA with PCV2-specific antibody confirmed that the
molecular DNA clone is infectious in vitro and that about 10-15% of the PK-15 cells were
transfected. PCV2-specific antigen was visualized by IFA in the nucleus, and to a lesser
degree, cytoplasm of the transfected cells (Fig. 2). The cells mock-transfected with the empty
pSK vector remained negative for PCV2 antigen.
EXAMPLE 3
In Vitro Transfection with the PCV2 Molecular DNA Clone and Generation of a Biologically
Pure and Homogenous PCV2 Infectious Virus Stock
To test the infectivity of the molecular DNA clone in vitro, PK-15 cells free of PCV1
contamination were grown in 8-well LabTek chamber slides. When the PK-15 cells reached
about 85% confluency, cells were transfected with the molecular DNA clone using
Lipofectamine Plus Reagents according to the protocol supplied by the manufacturer (Life

Technologies, Inc). Mock-transfected cells with empty pSK vector were included as controls.
Three days after transfection, the cells were fixed with a solution containing 80% acetone
and 20% methanol at 4° C for 20 min., and an immunofluorescence assay using a PCV2-
specific rabbit polyclonal antisera was performed to determine the in vitro infectivity of the
molecular DNA clone (see below).
i
To generate a biologically pure and homogeneous PCV2 infectious virus stock for the
animal inoculation experiment, PK-15 cells free of PCV1 contamination were cultivated in
T-25 culture flasks and transfected with the PCV2 molecular DNA clone. PK-15 cells were
grown to about 85% confluency in T-25 flasks. The cells were washed once with sterile PBS
buffer before transfection. For each transfection reaction in a T-25 flask, 12 ug of the PCV2
plasmid DNA was mixed with 16 ul of Plus Reagent in 0.35 ml of MEM media. A flask of
mock-transfected cells with empty pSK vector was included as the negative control. After
incubation at room temperature for 15 min., 50 ul of Lipofectamine Reagent diluted in 0.35
ml of MEM media was added to the mixture and incubated at room temperature for another
15 min. The transfection mixture was then added to a T-25 flask of PK-15 cells containing
2.5 ml of fresh MEM. After incubation at 37° C for 3 hrs, the media was replaced with fresh
MEM media containing 2 % FBS and 1 X antibiotics. The transfected cells were harvested
at 3 days post transfection and stored at -80° C until use. The infectious titer of the virus
stock was determined by IFA (see below).
Basically, biologically pure and homogenous PCV2 infectious virus stock was
generated by transfection of PK-15 cells with the PCV2 molecular DNA clone. PCV2 virions
produced by in vitro transfection were infectious since the transfected cell lysates were
successfully used to infect PK-15 cells. Thus, the PCV2 molecular DNA clone is capable of
producing infectious PCV2 virions when transfected in vitro. The infectious titer of the
homogenous PCV2 virus stock prepared from transfected cells was determined to be 1 x 104"5
TCID50/ml. This virus stock was used to inoculate pigs in Group 2. Lysates of cells mock-
transfected with the empty pSK vector were unable to infect PK-15 cells.

EXAMPLE 4
Virus Titration bv Immunofluorescence Assay (IFA)
To determine the infectious titer of the homogenous PCV2 virus stock, PK-15 cells
were cultivated on 8-well LabTek chamber slides. The virus stock was serially diluted 10-
fold in MEM, and each dilution was inoculated onto 10 wells of the monolayers of the PK-15
cells growing on the LabTek chamber slides. Wells of non-inoculated cells were included as
controls. The infected cells were fixed at 3 days post inoculation with a solution containing
80% acetone and 20% methanol at 4°C for 20 min. After washing the cells with PBS buffer,
the infected cells were incubated with a 1:1,000 diluted PCV2-specific rabbit polyclonal
antibody (S. D. Sorden et al., "Development of a polyclonal-antibody-based
immunohistochemical method for the detection of type 2 porcine circovirus in formalin-
fixed, paraffin-embedded tissue," J. Vet. Diagn. Invest. 11:528-530 (1999)) at 37°C for 1 hr.
The cells were then washed three times with PBS buffer, and incubated with a secondary
FITC-labeled goat anti-rabbit IgG (Kirkegaard & Perry Laboratories Inc, Gaithersburg, MD)
at 37°C for 45 min. After washing the slides three times with PBS buffer, and the slides
were mounted with fluoromount-G, cover-slipped and examined under a fluorescence
microscope. The 50% tissue culture infectious dose per ml (TCID50/ml) was calculated.
Initially, cells were transfected with a plasmid construct containing a single copy of PCV2
genome but the infectious PCV2 titer from the single genome construct is much lower than
the one containing the tandem genome. Therefore, the plasmid construct containing the
dimeric form of PCV2 genome was used for the in vitro and in vivo transfection experiments.
EXAMPLE 5
In Vivo Transfection of Pigs With the PCV2 Molecular DNA Clone and Experimental
Inoculation of Pigs With the Homogeneous PCV2 Infectious Virus Stock
Forty specific-pathogen-free (SPF) swine of 4 weeks of age were randomly assigned
into 4 rooms of 10 animals each. Prior to inoculation, the SPF pigs were tested for antibodies
to PCV, PRRSV, PPV and swine hepatitis E virus. Pigs in Group 1 were uninoculated and
served as negative controls. Pigs in Group 2 were each inoculated intranasally with about 1.9
x 105 TCID50 of the PCV2 infectious virus stock derived from the PCV2 molecular DNA
clone. Pigs in Group 3 received direct intrahepatic injection of the recombinant plasmid DNA

of the PCV2 molecular clone. Each pig was injected with a total of 200 ug of recombinant
plasmid DNA (the cloned PCV2 plasmid DNA), through an ultrasound-guided technique,
into 6 different sites of the liver. Pigs in Group 4 were each injected with a total of 200 µg of
the recombinant PCV2 plasmid DNA directly into the superficial iliac lymph nodes, and each
lymph node received two separate injections. The animals were monitored daily for clinical
signs of disease. Serum samples were collected from each animal at 0, 7, 14, 21, 28, 35 days
post inoculation (DPI). At 21 DPI, five pigs were randomly selected from each group and
necropsied. The remaining five animals in each group were necropsied at 35 DPI. Various
tissues and organs were collected during necropsy and processed for histological examination
and immunohistochemical staining (see below).
The results are shown in Table 1 below. All inoculated pigs from Groups 2, 3 and 4
were negative for PCV2 antibodies at 0 DPI. Two pigs in the uninoculated control Group 1
had detectable PCV2 maternal antibody at 0 DPI. The maternal antibody in these two piglets
waned by 7 DPI. No seroconversion to PCV2 antibody was detected in any of the 10
uninoculated control pigs. In Group 2 pigs intranasally inoculated with PCV2 infectious
virus, 1 piglet seroconverted to PCV2 antibody at 21 DPI. By 35 DPI, 4 of the 5 remaining
Group 2 pigs had seroconverted. Seroconversion in transfected animals from Groups 3 and 4
first appeared at 28 DPI. By 35 DPI, 5 of 5 remaining pigs from Group 3 and 3 of 5
remaining pigs from Group 4 had seroconverted to PCV2 antibody.
PPV antibodies were tested at 3 and 21 DPI for all pigs, and at 35 DPI for the
remaining pigs. Maternal antibodies to the ubiquitous swine agent PPV were detected in the
SPF piglets. The PPV HI antibody titers in all piglets but one decreased significantly from 3
DPI (an average titer of 1:2,665) to 21 DPI (an average titer of 1:246), indicating the
antibody detected in these piglets was passively derived. One piglet had a slightly increased
PPV HI titer from 1:32 at 3 DPI to 1:64 at 21 DPI, which is likely due to testing variation.
Serum samples collected from all pigs at 0, 21, and 35 DPI were further tested for PPV DNA
with a published PCR assay (J. M. Soucie el al., "Investigation of porcine parvovirus among
persons with hemophilia receiving Hyate: C porcine factor VIII concentrate," Transfusion
40:708-711 (2000)). No PPV viremia was detected from any pigs at any DPI, further
indicating the pigs were not infected by PPV.

Table 1. Seroconversion to PCV2 Specific Antibodies in Pigs Inoculated With PCV2
Live Virus or Directly Injected With Cloned PCV2 Plasmid DNA


EXAMPLE 6
PCR-RFLP Analyses
To measure PCV2 viremia in pigs transfected with PCV2 molecular DNA clone and
in pigs infected with PCV2 infectious virus stock, serum samples collected at different DPIs
were tested for the presence of PCV2 DNA by the general methods of a PCR-RFLP assay
previously described (M. Fenaux et al., 2000, supra). Viral DNA was extracted from 50 µl
of each serum sample using the DNAzol® reagent according to the protocol supplied by the
manufacturer (Molecular Research Center, Cincinnati, OH). The extracted DNA was
resuspended in DNase-, RNase-, and proteinase-free water and tested for PCV2 DNA by
PCR-RFLP (id). PCR products from selected animals were sequenced to verify the origin of
the virus infecting pigs.
Serum samples were collected from all control and inoculated animals at 0, 7, 14, 21,
28, and 35 DPIs and assayed for PCV2 viremia by detection of PCV2 DNA (id). The results
are shown in Table 2 below. PCV2 DNA was not detected in the Group 1 uninoculated
control pigs at any DPI. Viremia was detected in 7/10 pigs from Group 2 at 14 DPI and 8/10
by 35 DPI. Viremia lasted only a few weeks as the PCV2 DNA was not detectable at 28 DPI
and 35 DPI in all 5 remaining pigs from Group 2. In Group 3 pigs that were intrahepatically
injected with PCV2 molecular DNA clone, 8/10 pigs were viremic at 14 DPI, and 9/10 pigs
had detectable viremia by 35 DPI. Group 4 pigs were injected with PCV2 molecular DNA
clone into the lymph nodes. Two of 10 pigs at 14 DPI and 8 of 10 pigs at 21 DPI from
Group 4 were viremic. The results show that PCV2 molecular DNA clone is infectious when
injected directly into the liver and superficial iliac lymph nodes of SPF pigs. PCR products
amplified from selected animals were sequenced. The sequence of the PCR products
amplified from selected animals was identical to the corresponding region of the PCV2
molecular DNA clone.

Table 2. Detection of Viremia (PCV2 DNA) by PCR in Sera
of Inoculated and Control Pigs


EXAMPLE 7
Clinical Evaluation
Pigs were weighed on 0 DPI and at the time of necropsy. Rectal temperatures and
clinical respiratory disease scores, ranging from 0 to 6 (0 = normal, 6 = severe) (P. G. Halbur
et ah, "Comparison of the pathogenicity of two U.S. porcine reproductive and respiratory
syndrome virus isolates with that of the Lelystad virus," Vet. Pathol. 32:648-660 (1995)),
were recorded every other day from 0 to 35 DPI. Clinical observations including evidence of
central nervous system disease, liver disease (icterus), musculoskeletal disease, and changes
in body condition, were also recorded daily.
To evaluate the gross pathology and histopathology, five pigs from each group were
randomly selected for necropsies at 21 and 35 DPI. The necropsy team was blinded to
infection status of the pigs at necropsy. Complete necropsies were performed on all pigs. An
estimated percentage of the lung with grossly visible pneumonia was recorded for each pig
based on a previously described scoring system (id.). The scoring system is based on the
approximate volume that each lung lobe contributes to the entire lung: the right cranial lobe,
right middle lobe, cranial part of the left cranial lobe, and the caudal part of the left cranial
lobe each contribute 10% of the total lung volume, the accessory lobe contributes 5%, and
the right and left caudal lobes each contribute 27.5%. Other lesions such as enlargement of
lymph nodes were noted separately. Sections for histopathologic examination were taken
from nasal turbinate, lungs (seven sections) (id), heart, brain, lymph nodes
(tracheobronchial, iliac, mesenteric, subinguinal), tonsil, thymus, liver, gall bladder, spleen,
joints, small intestine, colon, pancreas, and kidney. The tissues were examined in a blinded
fashion and given a subjective score for severity of lung, lymph node, and liver lesions.
Lung scores ranged from 0 (normal) to 3 (severe lymphohistiocytic interstitial pneumonia).
Liver scores ranged from 0 (normal) to 3 (severe lymphohistiocytic hepatitis). Lymph node
scores were for an estimated amount of lymphoid depletion of follicles ranging from 0
(normal or no lymphoid depletion) to 3 (severe lymphoid depletion and histiocytic
replacement of follicles).
The serology protocol involved collecting blood on arrival at 11 to 12 days of age,
and from all pigs at 0, 7, 14, 21, 28, and 35 DPIs. Serum antibodies to PRRSV were assayed
using Herd Check PRRSV ELISA (IDEXX Laboratories, Westbrook, MA). Serum

antibodies to PPV were detected by a hemagglutination inhibition (HI) assay (H. S. Joo et al.,>
"A standardized haemagglutination inhibition test for porcine parvovirus antibody," Aust.
Vet. J. 52:422-424 (1976)). Serum antibodies to PCV2 were detected by a modified indirect
ELISA based on the recombinant ORF2 protein of PCV2 (P. Nawagitgul et al., "Modified
indirect porcine circovirus (PCV) type 2-based and recombinant capsid protein (ORF2)-
based ELISA for the detection of antibodies to PCV," Immunol. Clin. Diagn. Lab Immunol.
1:33-40 (2002)). A partially purified PCV2 antigen was prepared from Hi Five cells
(Invitrogen, Carlsbad, CA) infected with recombinant baculovirus containing the major
capsid ORF2 protein of PCV2 (P. Nawagitgul et al., "Open reading frame 2 of porcine
circovirus type 2 encodes a major capsid protein," J. Gen. Virol. 81:2281-2287 (2000)).
Cell lysates of Hi Five cells infected with wild-type baculovirus were prepared similarly and
served as negative control antigen. The Immulon 2 HB polystyrene microtiter plates (Dynex
Technologies Inc, Chantilly, VA) were coated with optimal concentrations of positive and
negative antigens at 4°C for 36 hrs. One hundred µl of each serum sample diluted 1:100 in
5% milk diluent (Kirkegaard & Perry Laboratories, Inc.) was added into each well. The
serum samples were tested in quadruplicate: 2 wells for negative control antigen and 2
parallel wells for PCV2 antigen. Positive control and negative control sera were included in
each plate. The sera were incubated at 37°C for 30 min and then washed 5 times with 0.1 M
PBS buffer containing 0.1% Tween-20. A peroxidase-labeled secondary anti-swine IgG
(Sigma Co, St. Louis, MO) was incubated at 37°C for 30 min. The plates were washed again
and incubated with 2,2"-azino-di-(3-ethylbenzthiazoline-6-sulfonate) (Kirkegaard & Perry
Laboratories Inc) at 37°C for 15 min. for color development. The optical density (OD) was
read at 405 nm. The corrected OD of each tested and control sera was calculated by
subtraction of mean OD value of the wells containing negative antigen from that of the
parallel wells containing PCV2 antigen. The data was normalized by dividing the corrected
OD value of a tested serum sample (S) with that of the positive control serum (P) and
reported as S/P ratios. The samples with S/P ratios 0.2 were
considered as negative, equivocal and positive, respectively.
From the results of the clinical evaluation, none of the control and inoculated pigs
showed obvious signs of disease resembling those of clinical PMWS. There was no
difference in weight gain or mean rectal temperatures between any of the four groups. The

control pigs of Group 1 remained normal throughout the experiment. There was mild
transient respiratory disease observed in the majority of the pigs in PCV2 DNA-transfected
and PCV2 virus-infected groups from 8 to 14 DPI. This was characterized by mild dyspnea
(clinical respiratory scores of 1 to 2) of one-to-two days duration in individual pigs and 5-6
days duration for the group.
There were no gross lesions observed in the control pigs at necropsy. Pigs in the three
inoculated groups had gross lesions limited to the lungs and lymph nodes (see Table 3,
below). The lesions were similar among pigs in the PCV2 plasmid DNA-transfected and
PCV2 virus-infected groups. Lungs failed to collapse and had random, multifocal,
moderately well-demarcated areas of tan-to-purple consolidation involving 0-2% of the lung
(Fig. 3) at 21 DPI, and 0-13% of the lung at 35 DPI. Lymph nodes were systemically
enlarged 2 to 5 times normal size, firm, and tan (Fig. 3) at both 21 and 35 DPI in most of the
pigs from all three PCV2-inoculated groups.
Microscopic examination revealed no lesions in any tissues of the control pigs except
for the livers. Eight of ten control pigs had very mild multifocal lymphoplasmacytic
inflammation predominately in the periportal regions of the liver as is commonly observed in
normal pigs and considered normal background (P. G. Halbur et al., 2001, supra).
Pigs from the two PCV2 plasmid DNA-transfected groups (intrahepatic and
intralymphoid) and the PCV2 virus-infected group (intranasal) had similar lesions in brain,
lung, heart, kidney, lymphoid tissues (tonsil, lymph nodes, spleen), ileum, and liver (see
Table 4, below). Brain lesions were observed in 23/30 of the pigs from the three inoculated
groups and were characterized as mild-to-moderate multifocal lymphoplasmacytic
meningoencephalitis with perivascular cuffing and gliosis. Lung lesions were observed in
28/30 PCV2-inoculated pigs and characterized as mild-to-moderate peribronchiolar lympho-
plasmacytic and histiocytic bronchointerstitial pneumonia (Fig. 3C). One pig from the PCV2
virus-infected Group 2 necropsied at 21 DPL and one pig each from the two PCV2 plasmid
DNA-transfected groups necropsied at 35 DPI had ulcerative and proliferative bronchiolitis
with fibroplasia and granulomatous inflammation in the lamina propria and peribronchiolar
regions of bronchi. Mild multifocal lymphoplasmacytic myocarditis was also observed in
18/30 PCV2-inoculated pigs. In 14/30 of the PCV2-inoculated pigs, mild-to-moderate
multifocal lymphoplasmacytic interstitial nephritis was observed. No lesions were observed

in the thymuses.. Mild-to-moderate lymphoid depletion (Fig. 4B) and histiocytic replacement
of follicles was observed in the tonsil of 8/30, in the spleen of 7/30, and in the lymph nodes
of 26/30 of the PCV2-inoculated pigs. Moderate granulomatous lymphadenitis with giant
cells (Fig. 4C) was observed at 21 DPI in three pigs inoculated intranasally with PCV2 virus,
and in one pig at 35 DPI in each of the PCV2 plasmid DNA-transfected groups. Mild
lymphoplasmacytic and histiocytic enterocolitis were observed in 3/5 pigs in the PCV2 virus-
infected group, in 3/5 pigs in the PCV2 plasmid DNA intrahepatically-transfected group, and
1/5 pigs in the PCV2 plasmid DNA intralymphoid-transfected group at 35 DPI. One pig in
each of the PCV2 plasmid DNA-transfected groups had mild lymphoid depletion with
histiocytic replacement and low numbers of giant cells in the Peyer"s patches. Mild-to-
moderate lymphoplasmacytic hepatitis was observed in 29/30 of the three PCV2-inoculated
pigs. Low numbers of widely scattered individually necrotic hepatocytes surrounded by
lymphohistiocytic inflammation was observed in one pig in each of the PCV2 plasmid DNA-
transfected groups at 21 DPI. Lesions in other tissues were unremarkable.
Microscopic lesions in the lung, liver and lymph nodes were scored according to
published scoring systems (Table 4, below) (P. G. Halbur et al, 2001, supra; P. G. Halbur et
al, 1995, supra). There were no acceptable scoring systems for other tissues and organs.
The average scores of lesions in lung and lymph nodes in pigs of the three PCV2-inoculated
groups were statistically different from those in the control pigs of Group 1. The average
scores of the liver lesions in pigs of the three PCV2-inoculated groups are not statistically
different from those of control pigs.





EXAMPLE 8
Immunohistochemistry
Immunohistochemistry (IHC) detection of PCV2-specific antigen was performed on
all tissues collected during necropsies at DPIs 21 and 35. A rabbit polyclonal PCV2-specific
antiserum was used for the IHC, and the general procedures have been previously described
(S. D. Sorden et al., 1999, supra).
For the detection and tissue distribution of PCV2 antigen, IHC staining of PCV2
antigen was done on brain, lungs, turbinate, heart, kidneys, tonsil, lymph nodes, spleen,
thymus, ileum, liver, gall bladder and pancreas of all pigs necropsied at 21 and 35 DPI. All
tissues from the control pigs were negative for PCV2 antigen. Tissue distribution of PCV2
antigen in the three PCV2-inoculated groups was similar (see Table 5, below). In the brain,
the PCV2 antigen was found predominately in mononuclear cells, fibroblast-like cells, and
endothelial cells in the meninges and choroid plexus and less often in endothelial cells and
perivascular mononuclear cells in the cerebrum and cerebellum. In the lungs, PCV2 antigen
was detected within alveolar and septal macrophages and in fibroblast-like cells in the lamina
propria of airways (Fig. 3D). In the heart, PCV2 antigen was detected in widely scattered
macrophages and endothelial cells. In kidneys, PCV2 antigen was detected within tubular
epithelial cells and mononuclear cells in the interstitium. In the lymphoid tissues (lymph
nodes, spleen, tonsil, and Peyer"s patches), PCV2 antigen was detected primarily within
macrophages and dendritic-like cells and giant cells within follicles (Fig. 4D). PCV2 antigen
was also detected within macrophages in the lamina propria of the small intestine. In the
liver, PCV2 antigen was detected within mononuclear cells and Kupffer cells. PCV2 antigen
was not detected in turbinate, thymus, or gall bladder.



EXAMPLE 9
Construction of the Nonpathogenic PCVI Infectious DNA Clone
The procedure used to construct a PCV1 infectious DNA clone is essentially the same
as that described herein for PCV2. A pair of PCR primers, KPNPCV1.U set forth in SEQ ID
NO:7 and KPNPCV1.L set forth in SEQ ID NO:8 (see Table 6, below), was designed based
on the published sequence of PCV1. This pair of primers amplifies the complete genome of
PCV1 with an overlapping region containing the unique KpnI restriction enzyme site. The
DNA of the PCVI virus was extracted from the contaminated. ATCC PK-15 cell line that was
obtained from the American Type Culture Collection (ATCC accession number CCL-33).
The PCV1 DNA was extracted from the ATCC PK-15 cells persistently infected with PCV1,
using the QIAmp DNA minikit (Qiagen, Inc.,Valencia, CA.). The extracted DNA was
amplified by PCR with AmpliTaq Gold Polymerase (Perkin-Elmer, Norwalk, CT). The PCR
cycles consisted of an initial step of 95°C for 10 min., followed by 35 cycles of denaturation
at 94°C for 1 min., annealing at 48°C for 1 min., extension at 72°C for 2 min., and a final
extension at 72°C for 7 min. The PCR product of expected size was separated by gel
electrophoresis and purified by the glassmilk procedure using a Geneclean Kit (Bio 101, Inc.,
La Jolla, CA). The purified PCR product containing the complete PCV1 genome was first
ligated into the advanTAge plasmid vector (Clontech, Palo Alto, CA). Escherichia coli
DH5? competent cells were used for transformation. The recombinant plasmids were
verified by restriction enzyme digestion. The full length PCV1 genomic DNA was excised
from the advanTAge vector by digestion with Kpnl restriction enzyme. The full-length
PCV1 genomic DNA was ligated into pBluescript SK(+) (pSK) vector (Stratagene, La Jolla,
CA) with T4 DNA ligase at 37°C overnight. Recombinant plasmids containing the full-
length PCVI genome were isolated with a Qiagen plasmid mini kit (Qiagen, Valencia, CA)
and were verified by restriction enzyme digestion and DNA sequencing. The full-length
PCV1 genomic DNA was excised from the pSK vector by Kpnl digestion, and dimmerized to
make the PCV1 infectious DNA clone as described above in Example 2 for the PCV2
infectious clone. These tandem dimers were made because the dimmerized tandem DNA
clones are advantageously found to be more efficient to transfect cells and produce infectious
virions. To make the tandem dimer of the PCV1 DNA, the digested PCV1 genomic DNA
was ligated with T4 DNA ligase at 37°C for only 10 min., which favors the production of

tandem dimers. The tandem dimers were subsequently cloned into pBluescript SK(+) (pSK)
vector (Stratagene, La Jolla, CA). Recombinant plasmids containing tandem dimers of PCV1
genome (herein referred to as "PCV1 DNA clone") were confirmed by PCR, restriction
enzyme digestion, and DNA sequencing. The DNA concentration of the recombinant
plasmids was determined spectrophotometrically.

-49"



EXAMPLE 10
Evaluation of Infectivitv of the PCV1 DNA Clone When
Transfected into PK-15 Cells Free of Virus Contamination
The infectivity of the PCV1 molecular DNA clone was determined by in vitro
transfection of the PK-15 cells. IFA with PCV1 specific monoclonal antibody (a gift from
Dr. Gordon Allan, Belfast, U.K.) confirmed that the PCV1 molecular DNA clone is
infectious in PK-15 cells. PCVl-specific antigen was visualized by IFA in the nucleus, and
to a lesser degree cytoplasm of the transfected cells. The cells mock-transfected with the
empty pSK vector remained negative for PCV1 antigen.
EXAMPLE 11
Construction of a Chimeric PCV1-2 Viral DNA Clone
A chimeric virus was constructed between the nonpathogenic PCV1 and the PMWS-
associated PCV2 by using infectious DNA clones of PCV1 and PCV2. To construct a
chimeric PCV1-2 DNA clone, the ORF2 capsid gene of the nonpathogenic PCV1 was
removed from the PCV1 infectious DNA clone, and replaced with the immunogenic ORF2
capsid gene of the pathogenic PCV2 in the genome backbone of PCV1 (see Figs. 5 and 6).
Two pairs of PCR primers were designed. The first primer pair for PCV2 ORF2, Psi 1-5 set
forth in SEQ ID NO: 11 and Acl 1-6 set forth in SEQ ID NO: 12, was designed with point
mutations at the 5" ends of the primers to create restriction enzyme sites AclI and PsiI to
amplify the ORF2 gene of PCV2 and introduce flanking PsiI and AclI restriction enzyme
sites by point mutation. The PCR reaction for the PCV2 ORF2 amplification consisted of an
initial step at 95°C for 9 min., followed by 38 cycles of denaturation at 95°C for 1 min.,
annealing at 48°C for 1 min., extension at 72°C for 1 min., and a final extension at 72°C for 7
min.
A second pair of PCR primers, Hpa 1-2 set forth in SEQ ID NO:9 and Nar 1-3 set
forth in SEQ ID NO: 10, was designed for the amplification of the pSK+ vector and its PCV1
genome insert. Point mutations were introduced at the 5" ends of the PCR primers to create
flanking restriction enzyme sites NarI and HpaI. This primer pair amplified the pSK+ vector
and its insert PCVI genomic DNA lacking the ORF2 capsid gene, that is, the PCVI genome
minus the PCV1 ORF2 (pSK-PCVl AORF2) by using the PCV1 infectious DNA clone as

the PCR template. The PCR reaction consisted of an initial step at 95°C for 9 min., followed
by 38 cycles of denaturation at 95°C for 1 min., annealing at 50°C for 1min., extension at
72°C for 3.5 min., and a final extension at 72°C for 7min. The PCV2 ORF2 PCR product
was digested with the AclI and PsiI to remove the introduced point mutations. The pSK-
PCV1 AORF2 product (the pSK vector-PCVl genome PCR product lacking 0RF2 gene of
PCVI) was digested with the NarI and HpaI to remove the PCR introduced point mutations.
The latter digestion produced a sticky end and a blunt end complementary to the PCV2
ORF2 PCR product digested by the AclI and PsiI restriction enzymes. The digested PCV2
ORF2 product and the ORF2-deleted pSK-PCVl product were ligated with T4 DNA ligase
to form the chimeric PCV1-2 genomic DNA clone, in which the ORF2 gene of PCV1 is
replaced with the ORF2 gene of PCV2. Once the two PCR products were digested and
religated, all the PCR introduced point mutations used to facilitate cloning were removed in
the resulting chimeric clone. Escherichia coli DH5? competent cells were transformed. The
recombinant plasmids containing the chimeric DNA clone were isolated and confirmed by
PCR, restriction enzyme digestion and partial DNA sequencing. The full-length chimeric
PCV1-2 genome was excised from the pSK+ vector (the recombinant plasmid) with KpnI
digestion. The chimeric DNA genome was then dimmerized by a short 10-minute ligation
reaction with T4 DNA ligase that favors the formation of linear dimers to produce the PCV1-
2 chimeric infectious DNA clone (Fig. 6). The recombinant plasmids containing two copies
of the chimeric viral genome were confirmed by PCR, restriction enzyme digestion and DNA
sequencing.
EXAMPLE 12
Evaluation of In Vitro Infectivitv of PCV1-2 Chimeric DNA Clone
The viability of the chimeric PCV DNA clone (nonpathogenic PCV1 with the
immunogenic capsid gene of PCV2) was tested in PK-15 cells. When PK-15 cells were
transfected with the chimeric viral DNA clone, viral antigen specific for PCV2 ORF2 capsid
was detected by IFA at about 2 days post-transfection. The PCV1 capsid antigen was not
detected in transfected cells. This experiment indicated that the chimeric DNA clone is
infectious in vitro, is replicating in PK-15 cells and producing the immunogenic capsid
protein of PCV2.

EXAMPLE 13
Construction of a Reciprocal Chimeric PCV2-1 DNA Clone
To construct a reciprocal PCV2-1 chimeric DNA clone, the ORF2 capsid gene of
PCV2 is replaced by that of the non-pathogenic PCV1 in the genome backbone of the
pathogenic PCV2 (Fig. 6). Two PCR primer pairs were designed: the pair, Bgl-II-ORF2 set
forth in SEQ ID NO: 13 and SpH-I-ORF2 set forth in SEQ ID NO: 14, amplifies the PCV1
ORF2 gene and introduces flanking BglII and SpHI restriction enzyme sites by point
mutation. The second PCR primer pair, Bgl-II-PCV2 set forth in SEQ ID NO: 15 and SpH-I-
PCV2 set forth in SEQ ID NO: 16, amplified the pSK vector and the PCV2 genome minus the
ORF2 gene (pSK-PCV2 AORF2) by using the PCV2 infectious DNA clone as the PCR
template, and introduced flanking restriction enzymes sites BglII and SpHI by point mutation.
The pSK-PCV2 AORF2 product and the PCV1 ORF2 PCR product were digested by BglII
and SpHI restriction enzymes to produce complementary sticky and blunt ends ligated
together. After transformation into E. Coli cells, the authentic recombinant plasmids were
isolated and confirmed by enzyme digestion and partial DNA sequencing. The full-length
reciprocal chimeric PCV2-1 genome was excised from the recombinant plasmid by SacII
digestion, and dimmerized as described herein to produce the reciprocal chimeric PCV2-1
infectious clone.
EXAMPLE 14
In vitro Transfection of PK-15 Cells with PCV1, PCV2,
PCV1-2 and PCV2-1 DNA Clones
The infectivity of PCV2 clone in vitro and in vivo has been demonstrated in the above
Examples 3-5. To test the infectivity of the PCV1 and two chimeric clones in vitro, PK-15
cells free of PCV1 contamination prepared per the method of Example 1 were grown in 8-
well LabTek chambers slides (Nalge Nunc Int., Denmark). When the PK-15 cells reached
about 80% confluence cells were transfected with PCV1, PCV2, PCV1-2 and PCV2-1 DNA
clones respectively, using the Lipofectamine Plus Reagent according to the protocols
supplied by the manufacturer (Life Technologies, Inc.). Mock-transfected cells with empty
pSK vector were included as controls. Three days after transfection, the cells were fixed

with a solution containing 80% acetone and 20% methanol at 4°C for 20 min. Evidence of
infectivity and virus replication in cells transfected with the PCVI and PCV2-1 DNA clones
were confirmed by indirect immunofluorescence assay (IFA) using monoclonal antibody
against PCV1 ORF2 capsid gene, kindly provided by Dr. G. M. Allan (G. M. Allan et at.,
"Production, preliminary characterization and applications of monoclonal antibodies to
porcine circovirus," Vet. Immunol. Immunopathol. 43:357-371 (1994)). The fixed cells were
washed with phosphate buffered saline (PBS) and incubated with 1:20 diluted PCV1
monoclonal antibody at 37°C for 1 hour. The cells were then washed three times with PBS
buffer and incubated with fluorescein isothiocyanate (FITC) labeled goat anti-mouse
immunoglobulin G (Kirkegaard & Perry Laboratories, Inc., Gaithersburg, Md.) at 37°C for
45 min. After washing three times with PBS buffer, the slides were mounted with
fluoromount-G, coverslipped, and examined under a fluorescence microscope. The
infectivity of cells transfected with the PCV2 and the chimeric PCV1-2 DNA clones were
confirmed by IFA using antibody specific for PCV2, as previously described in Example 4.
The infectivity of the PCV1, the chimeric PCV1-2 DNA and the reciprocal chimeric
PCV2-1 DNA clones were substantiated by the in vitro transfection of PK-15 cells. Two
identical copies of the complete PCVI genome were ligated in tandem into the pSK vector to
produce the PCV1 DNA clone (Fig. 6). The chimeric PCV1-2 DNA clone had the ORF2
capsid gene of PCVI replaced by that of the pathogenic PCV2 in the backbone of the
nonpathogenic PCV1 genome. The reciprocal chimeric PCV2-1 DNA clone had the ORF2
capsid gene of PCV2 replaced by that of the nonpathogenic PCVI in the backbone of the
pathogenic PCV2 genome. If infectious in vitro, the chimeric PCV1-2 DNA clone will
produce the ORF2 capsid antigen of PCV2 and the reciprocal chimeric PCV2-1 DNA clone
will express PCV1 ORF2 capsid antigen in transfected PK-15 cells. The results showed that
the PCV1, the chimeric PCV1-2 and the reciprocal chimeric PCV2-1 DNA clones were all
surprisingly shown to be infectious when transfected into PK-15 cells and expressed
respective viral capsid antigen proteins as demonstrated by IFA using antibodies specific for
PCV1 or PCV2. IFA using monoclonal antibodies against PCV1 ORF2 and antibodies
against PCV2 confirmed that the PCV1 DNA and the PCV1-2 DNA clones were infectious.
IFA using PCV1 ORF2-specific monoclonal antibody showed that the PCV1-2 chimeric
DNA clone was also infectious. About 10-20% of the transfected PK-15 cells were positive

for PCV1 capsid antigen and PCV2 antigen, and expressed PCV1 ORF2 antigen, within the
nucleus of transfected cells (Fig. 7).
EXAMPLE 15
Experimental Inoculation of Pigs with PCV1, PCV2. Chimeric
PCVI-2 and Reciprocal Chimeric PCV2-1 DNA Clones
To evaluate the immunogenicity and pathogenicity of the chimeric DNA clones, forty
specific-pathogen-free (SPF) pigs of 4-6 weeks of age were randomly assigned into five
rooms of 8 animals each. Prior to inoculation, animals were tested for antibodies to PCV,
PRRSV, PPV, and swine hepatitis E virus. In addition, pre-inoculation serum samples were
tested by PCR for PCV1 and PCV2 nucleic acid to confirm that the pigs are not naturally
infected by either of the viruses. The PCV1, PCV2, PCVI-2 and PCV2-1 DNA clones were
all inoculated by direct injection of the cloned plasmid DNA into the superficial iliac lymph
nodes of pigs. Pigs in Group 1 received phosphate buffered saline (PBS buffer) and served
as the negative control. Group 2 pigs were each injected into the superficial iliac lymph
nodes with 200 µg of infectious PCV1 DNA clone. Group 3 pigs were each injected with
200 |ig of infectious PCV2 DNA clone. Group 4 pigs each received injections of 200 µg of
infectious chimeric PCV1-2 DNA clone. Group 5 pigs each received 200 µg of the
infectious reciprocal chimeric PCV2-1 DNA clone. All animals were monitored daily for
clinical signs of disease. Serum samples were collected from each animal at -2, 7, 14, 21, 28,
35, 42 and 49 days post-inoculation (DPI). At 21 DPI, four randomly selected animals from
each group were necropsied. The remaining four animals in each group were necropsied at
49 DPI. Various tissues and organs were collected during necropsy as previously described in
Example 7, and processed for histological examination.
The immunogenicity of the PCV1, the PCV2 and the chimeric infectious DNA clones
was examined in the pigs. Serum samples collected from all control and inoculated animals
at -2 (0), 7, 14, 21, 28, 35, 42 and 49 DPIs were assayed for PCV1, PCV2, PCVI-2 and
PCV2-1 viremia by PCR detection of clone-specific DNA sequences, for anti-PCVI antibody
by IFA and for anti-PCV2 ORF2 antibody by ELISA. Prior to inoculation at -2 DPI, animals
from all five groups tested negative by PCR for both PCV1 and PCV2 DNA.

Negative control animals were negative for both PCV1 and PCV2 viremia throughout
the study (see Table 7, below). Five pigs in the uninoculated control group had detectable
PCV2 maternal antibody at -2 DPI and 2 pigs had detectable PCVI maternal antibodies at 7
DPI (see Table 8, below). The maternal antibodies to both PCVI and PCV2 in these piglets
waned by 21 DPI. No seroconversion to either PCVI or PCV2 was detected in any of the 8
uninoculated control pigs throughout the study.
In the PCV1 inoculated group, viremia was first detected in an inoculated pig at 7
DPI (Table 7, below), and was last detected at 35 DPI. Five out of 8 animals inoculated with
PCV1 infectious DNA clone were positive for PCV1 viremia. Average length of continuous
PCV1 viremia was 0.625 weeks. By 21 DPI, all animals in the PCV1 inoculated group had
seroconverted to PCV1 and remained positive to PCV1 antibodies until the end of the study
at 49 DPI.
The PCV2 DNA clone is shown herein to be infectious in pigs. In the PCV2 DNA
clone inoculated group, PCV2 viremia was first detected at 7 DPI (Table 7, below). By 21
DPI, all PCV2 inoculated Group 3 animals were positive for PCV2 viremia. The average
length of PCV2 viremia was 2.12 weeks. Two pigs in the PCV2 inoculated group had
detectable levels of maternal PCV2 antibodies at 7 DPI (Table 8, below), and the maternal
antibodies in these piglets waned by 14 DPI. Seroconversion to PCV2, assayed by a PCV2-
specific ELISA, was first detected at 35 DPI. By 42 DPI, all pigs inoculated with PCV2
infectious DNA clone had seroconverted to PCV2.
In Group 4 pigs inoculated with PCV1-2 chimeric infectious DNA clone, viremia
specific for the chimeric virus was first detected at 14 DPI (Table 7, below). Four out of 7
inoculated animals became viremic to PCV1-2 between 14 DPI and 42 DPI. The average
length of chimeric PCV1-2 viremia was 1 week. One pig had detectable levels of maternal
PCV2 antibodies at 7 and 14 DPI, but the maternal antibody waned by 21 DPI (Table 8,
below). Seroconversion to PCV2 ORF2-specific antibody first occurred at 28 DPI. By 49
DPI, all pigs inoculated with chimeric PCV1-2 DNA clone had seroconverted to PCV2
ORF2-specific antibody.
In pigs inoculated with the reciprocal chimeric PCV2-1 clone, viral DNA specific for
PCV2-1 chimeric virus was not detected in serum samples (Table 7, below). However, by 21
DPI all animals in Group 5 seroconverted to PCV1 antibody. PCR products amplified from

selected pigs in each group were sequenced and confirmed to be the authentic respective
infectious clones used in the inoculation in each group.




EXAMPLE 16
Clinical Evaluation
Pigs were weighed on 0 DPI and at the time of necropsies. Rectal temperatures and
clinical respiratory scores, ranging from 0 to 6 (0 = normal; 6 = severe) (P. G. Halbur et ah,
"Comparison of the pathogenicity of two U.S. porcine reproductive and respiratory syndrome
virus isolates with that of the Lelystad virus," Vet. Pathol. 32:648-660 (1995)), were recorded
every other day from 0 to 49 DPI. Clinical observations, including evidence of central
nervous system disease, liver disease (icterus), muscoloskeletal disease and changes in body
condition, were also recorded daily. A team of two people performed all clinical evaluations.
None of the control or inoculated pigs showed obvious signs of the full-spectrum
clinical PMWS. There were no differences in weight gain or mean rectal temperatures
between any of the groups. One of the pigs from PCV1-2 inoculated Group 3 died one day
after inoculation. After necropsy and clinical analysis, no pathogenic agent was detected and
death was not associated with the inoculation procedure or the chimeric PCV1-2 virus.
EXAMPLE 17
Gross Pathology and Histopathology
Four pigs from each group were necropsied at 21 and 49 DPI, respectively. The
necropsy team was blinded to infection status of the pigs at necropsy. Complete necropsies
were performed on all pigs. An estimated percentage of the lung with grossly visible
pneumonia was recorded for each pig based on a previously described scoring system (P. G.
Halbur et al., 1995, supra). Other lesions such as enlargement of lymph nodes were noted
separately. Sections for histopathologic examination were taken from nasal turbinate, lungs
(seven sections) (id), heart, brain, lymph nodes (tracheobronchial, iliac, mesenteric,
subinguinal), tonsil, thymus, liver, gall bladder, spleen, joints, small intestine, colon,
pancreas, and kidney. The tissues were examined in a blinded fashion and given a subjective
score for severity of lung, lymph node, and liver lesions as described in Example 7. Lung
scores ranged from 0 (normal) to 3 (severe lymphohistiocytic interstitial pneumonia). Liver
scores ranged from 0 (normal) to 3 (severe lymphohistiocytic hepatitis). Lymph node scores
were for an estimated amount of lymphoid depletion of follicles ranging from 0 (normal or

no lymphoid depletion) to 3 (severe lymphoid depletion and histiocytic replacement of
follicles).
To determine the pathogenicity of PCV1, PCV2, chimeric PCV1-2 and reciprocal
chimeric PCV2-1 infectious DNA clones in pigs, gross lesions were examined first. The
results are shown in Table 9 below. Lymph nodes of animals from the uninoculated control
Group 1 were normal at both 21 and 49 DPIs. Pigs in the four inoculated groups had variable
degrees of gross lesions limited to the lymph nodes. In PCV1 inoculated Group 2 pigs,
lymph nodes were grossly normal at 21 DPI, however, mild to moderate swelling and
discoloration of lymph nodes was detected at 49 DPI. All PCV2 inoculated Group 3 pigs had
enlarged lymph nodes two to five times the normal size, that were firm and tan colored at
both 21 and 49 DPIs. Lymph nodes from chimeric PCV1-2 inoculated animals were mild to
moderately swollen and discolored at both 21 and 49 DPIs in 5 out of 7 pigs. In Group 5
pigs, inoculated with the PCV2-1 clone, 1 out of 8 animals had mild swelling and
discoloration of the lymph nodes at 21 DPI. The average scores of gross lesions of the lymph
nodes in pigs inoculated with chimeric PCV1-2 clone were not statistically different from
those in Groups 1, 2, and 5, but were statistically different from those of the pathogenic
PCV2 inoculated Group 3 pigs at both 21 and 49 DPIs. Average lymphoid gross lesion scores
on 49 DPI from the PCV1, PCV2, and PCV1-2 inoculated animals were not statistically
different from each other, but were all statistically different from the average gross lesion
scores of Groups 1 and 5.
Next, microscopic lesions were examined. The results are shown in Table 10 below.
No microscopic lesions were detected in either uninoculated control Group 1 pigs or PCV1
inoculated Group 2 pigs at any DPI. Microscopic lung lesions characterized as mild
peribronchiolar lymphoplasmacytic and histiocytic bronchointerstitial pneumonia, were
observed in 1 out 8 of the PCV2 inoculated pigs. In PCV1-2 and PCV2-1 inoculated animals,
no microscopic lesions were observed in the lungs. No lesions were observed in the
thymuses of any inoculated pigs. Mild multifocal lymphoplasmacytic myocarditis was
observed in 2 of 8 pigs in the PCV2 inoculated group. Heart tissues from PCV1-2 and PCV2-
1 inoculated animals were free of microscopic lesions. Mild multifocal lymphoplasmacytic
interstitial nephritis was observed in 4 out of 8 pigs in PCV2 inoculated group, in 2 out of 7
PCV1-2 inoculated pigs and in 1 out of 8 PCV2-1 inoculated pigs. Mild-to-moderate

lymphoid depletion and histiocytic replacement of follicles were observed in the tonsil in 5
out of 8 pigs, in the spleen in 3 out of 8 pigs, and in the lymph nodes in 8 out of 8 pigs in the
PCV2-inoculated group. In the chimeric PCV1-2 inoculated animals, mild lymphoid
depletion and histiocytic replacement of follicles were observed in the lymph nodes of 2 out
of 7 pigs but were not detected in either the spleen or tonsils. No lymphoid depletion and
histiocytic replacement of follicles were observed in the lymph nodes, spleen or tonsils of the
reciprocal chimeric PCV2-1 inoculated animals. Mild-to-moderate lymphoplasmacytic
hepatitis was observed in 7 out of the 8 PCV2-inoculated pigs. Mild lymphoplasmacytic
hepatitis was observed in 2 out of the 7 chimeric PCV1-2 inoculated pigs. No
lymphoplasmacytic hepatitis was observed in reciprocal chimeric PCV2-1 inoculated pigs.
Lesions in other tissues were unremarkable.
Microscopic lesions in the lung, liver, and lymph nodes were scored according to a
published scoring system (P. G. Halbur et al., 1995, supra). The results are shown in Table
10 below. Average scores of lesions in lymph nodes in pigs from the chimeric PCV1-2
inoculated Group 4 were similar to those from Groups 1, 2 and 5 but were statistically
different from those of the pathogenic PCV2 inoculated Group 3 pigs, at both 21 and 49
DPIs. Average microscopic liver lesion scores from the chimeric PCV1-2 inoculated group
at 21 DPI were statistically different from those of PCV2 inoculated Group 3 animals but
were similar to those of Groups 1, 2 and 5 pigs at 21 DPI. At 49 DPI, the average
microscopic liver scores from Group 4 chimeric PCV1-2 inoculated pigs were not
statistically different from those of Groups 1, 2, 3 and 5 pigs. There were no acceptable
scoring systems for other tissues or organs.





EXAMPLE 18
Serology
Blood was collected from all pigs at -2, 7, 14, 21, 28, 35, 42 and 49 DPIs. Serum
antibodies to PRRSV were assayed using Herd Check PRRSV ELISA (IDEXX Laboratories,
Westbrook, MA). Serum antibodies to PPV were detected by a hemagglutination inhibition
(HI) assay (H. S. Joo et al., "A standardized haemagglutination inhibition test for porcine
parvovirus antibody," Aust. Vet. J. 52:422-424 (1976)). Serum antibodies to PCV2 were
detected by a modified indirect ELISA based on the recombinant 0RF2 capsid protein of
PCV2 as described hereinabove (see also P. Nawagitgul et al., "Modified indirect porcine
circovirus (PCV) type 2-based and recombinant capsid protein (ORF2)-based ELISA for the
detection of antibodies to PCV," Immunol. Clin. Diagn. Lab Immunol. 1:33-40 (2002)).
Serum antibodies to PCV1 were detected by an indirect immunofluorescence assay (IFA).
PK-15 cells infected with PCV1 were grown on eight-well LabTek chamber slides. When the
infected PK-15 cells reach about 95-100% confluency, the infected cells were fixed with a
solution containing 80% acetone and 20% methanol at 4°C for 20 min. The fixed cells were
washed once with PBS buffer. One hundred microliters of 1:10 diluted pig serum sample in
PBS was added to the chambers, and incubated for 1 hour at 37°C. The cells were then
washed three times with PBS and incubated for 45 min. at 37°C with FITC-labeled goat anti-
swine secondary antibody. The slides were subsequently washed three times with PBS,
mounted with fluoromount-G, coverslipped and examined under a fluorescent microscope.
For the positive control, PCV1 infected cells were incubated with a diluted PCV1 specific
monoclonal antibody (gift of Dr. G. M. Allan), followed by an incubation with FITC-labeled
goat anti-mouse IgG (Kirkegaard & Perry Laboratories, Inc., Gaithersburg, Md.). For the
negative control, PCV1 infected cells were incubated with 1:10 diluted swine serum free of
PCV1 and PCV2 antibody, followed by an incubation with FITC-labeled goat anti-swine IgG
(Kirkegaard& Perry Laboratories, Inc., Gaithersburg, Md.).
EXAMPLE 19
PCR Detection
To detect PCV1, PCV2, chimeric PCV1-2 and reciprocal chimeric PCV2-1 viremia in
sera from inoculated pigs, serum samples collected at different DPIs were tested by PCR.

Viral DNA was extracted from 100 µl of each serum sample using DNAzol reagent
according to the manufacturer"s protocol (Molecular Research Center, Cincinnati, OH). The
extracted DNA was resuspended in DNase, RNase and proteinase-free water. To amplify
clone-specific genomic sequences of PCV1, PCV2, chimeric PCV1-2 and chimeric
reciprocal PCV2-1, two sets of nested PCR primer pairs were designed (Table 6, above).
The first set of nested primers was designed based on published PCV1 sequences. Primers
Gen.PCVl set forth in SEQ ID NO:20 and Orf.PCVl set forth in SEQ ID NO: 19 amplified a
400 bp fragment in the presence of the PCV1 genome. The nested primers,
nested.Gen.PCVl set forth in SEQ ID NO:22 and nested.Orf.PCVl set forth in SEQ ID
NO:21, amplified a 220 bp fragment.
To detect PCV2 viremia, PCV2 primer pair Gen.PCV2 set forth in SEQ ID NO:24
and Orf.PCV2 set forth in SEQ ID NO:23 amplified a 900 bp fragment in the presence of
PCV2 in the first round of PCR. Primers nested.Gen.PCV2 set forth in SEQ ID NO:26 and
nested.Orf.PCV2 set forth in SEQ ID NO:25 amplified a 600 bp fragment in the nested PCR.
To detect chimeric PCV1-2 viremia, the first round of PCR reaction employed the
PCVl-specific primer Gen.PCVl set forth in SEQ ID NO:20 and the PCV2 ORF2-specific
primer Orf.PCV2 set forth in SEQ ID NO:23 to amplify a chimeric fragment of 580 bp. For
the nested PCR, PCVl-specific primer nested.Gen.PCVl set forth in SEQ ID NO:22 and the
PCV2 ORF2-specific primer nested.Orf.PCV2 set forth in SEQ ID NO:25 were used to
amplify a chimeric fragment of 370 bp.
To detect reciprocal chimeric PCV2-1 viremia, the first round of PCR employed the
PCV2-specific primer Gen.PCV2 set forth in SEQ ID NO:24 and the PCV1 ORF2-specific
primer Orf.PCVl set forth in SEQ ID NO: 19 to amplify a chimeric fragment of 700 bp. For
the nested PCR, the PCV2-specific primer nested.Gen.PCV2 set forth in SEQ ID NO:26 and
the PCV1 ORF2-specific primer nested.Orf.PCVl set forth in SEQ ID NO:21 were used to
amplify a 460 bp chimeric fragment. All PCR parameters were essentially the same,
consisting of 38 cycles of denaturation at 94°C for 1 min., annealing at 45°C for 1 min., and
extension at 72°C for 1.5 min. The serum samples from negative control pigs were tested by
a PCR-RFLP diagnostic assay, which can detect and differentiate both PCV1 and PCV2 as
described previously (M. Fenaux et al., "Genetic characterization of type 2 porcine circovirus
(PCV-2) from pigs with postweaning multisystemic wasting syndrome in different

geographic regions of North America and development of a differential PCR-restriction
fragment length polymorphism assay to detect and differentiate between infections with
PCV-1 and PCV-2," J. Clin. Microbiol. 38: 2494-503 (2000)). PCR products from selected
animals in each group were sequenced to verify the origin of the virus infecting pigs.
EXAMPLE 20
Immunohistochemistry (IHC)
IHC detection of PCV2-specific antigen was performed on lymph node tissues
collected from all pigs necropsied at 21 and 49 DPIs. A rabbit polyclonal antiserum against
PCV2 was used for the IHC, according to the general procedures described previously (S. D.
Sorden et al.t "Development of a polyclonal-antibody-based immunohistochemical method
for the detection of type 2 porcine circovirus in formalin-fixed, paraffin-embedded tissue," J.
Vet. Diagn. Invest. 11:528-530 (1999)).
Based on the IHC staining of PCV2-specific antigen, lymphoid tissues from the
uninoculated control, PCV1 and PCV2-1 inoculated pigs were negative for PCV2 antigen.
PCV2 antigen was detected in lymphoid tissues of 7 out of 8 animals in the PCV2 inoculated
group. PCV2 antigen was also detected in lymphoid tissue of 1 out of 7 pigs from the
chimeric PCV1-2 inoculated group.
In the foregoing, there has been provided a detailed description of particular
embodiments of the present invention for the purpose of illustration and not limitation. It is to
be understood that all other modifications, ramifications and equivalents obvious to those
having skill in the art based on this disclosure are intended to be included within the scope of the
invention as claimed.







WE CLAIM:
1. An infectious chimeric nucleic acid molecule of porcine circovirus (PCV1-2)
characterized by having a nucleic acid molecule encoding an infectious, nonpathogenic
PCV1 which contains an immunogenic open reading frame (ORF) gene of a pathogenic
PCV2 in place of an ORF gene of the PCV1 nucleic acid molecule.
2. The chimeric nucleic acid molecule as claimed in Claim 1, wherein the
immunogenic PCV2 ORF gene replaces the same ORF gene position in the PCV1 nucleic
acid molecule.
3. The chimeric nucleic acid molecule as claimed in Claim 2, wherein the
immunogenic ORF gene is the ORF2 capsid gene.
4. The chimeric nucleic acid molecule as claimed in Claim 3, wherein the chimeric
nucleic acid molecule has a nucleotide sequence set forth in SEQ ID NO:2, its
complementary strand or a nucleotide sequence which has at least 95% homology to the
nucleotide sequence of SEQ ID NO:2.
5. A biologically functional plasmid or viral vector containing the chimeric nucleic
acid molecule as claimed in Claim 4.
6. The plasmid as claimed in Claim 5 having ATCC Patent Deposit Designation PTA-
3912.
7. A microbial host cell transfected by a vector comprising the chimeric nucleic acid
molecule as claimed in Claim 4.
8. An avirulent, infectious chimeric porcine circovirus (PCV1-2) produced by a
biologically functional plasmid, a biologically functional viral vector or a microbial host cell
containing the chimeric nucleic acid molecule as claimed in Claim 4
9. The infectious chimeric porcine circovirus as claimed in Claim 8, wherein said cells
containing the chimeric nucleic acid molecule are contained in or derived from a plasmid
having ATCC Patent Deposit Designation PTA-3912.
10. A process for the production of an immunogenic polypeptide product, said process
comprising: growing, under microbial nutrient conditions, prokaryotic or eucaryotic host
cells transfected with a nucleic acid molecule as claimed in Claim 4 in a manner allowing

Expression of said polypeptide product, and isolating the desired polypeptide product of the
expression of said nucleic acid molecule.
11. An immunogenic polypeptide product of the expression as claimed in Claim 10.
12. An immunogenic polypeptide comprising the amino acid sequence set forth in SEQ
ID NO:4 or a biologically active variant thereof.
13. A viral vaccine that protects a pig against viral infection or postweaning
multisystemic wasting syndrome (PMWS) caused by PCV2 characterized by comprising a
nontoxic, physiologically acceptable carrier and an immunogenic amount of a member
selected from the group consisting of:
(a) a chimeric nucleic acid molecule having a nucleotide sequence set forth in SEQ
ID NO:2, its complementary strand or a nucleotide sequence having at least 95%
homology to the nucleotide sequence of SEQ ID NO:2;
(b) a biologically functional plasmid or viral vector containing the chimeric
nucleic acid molecule, the complementary strand or the nucleotide sequence having at least
95% homology to the nucleotide sequence of SEQ ID NO:2;
(c) an avirulent, infectious chimeric porcine circovirus made from a chimeric nucleic
acid molecule of PCV1-2;
(d) a polypeptide having the amino acid sequence set forth in SEQ ID NO:4 or a
biologically active variant thereof;
(e) an infectious PCV2 molecular DNA clone having ATCC Patent Deposit
Designation PTA-3913 or a PCV2 DNA clone derived therefrom; and
(f) a biologically functional plasmid or viral vector containing the infectious
PCV2 molecular DNA clone having ATCC Patent Deposit Designation PTA-3913 or the
PCV2 DNA clone derived therefrom.
14. The viral vaccine as claimed in Claim 13, wherein said vaccine contains live
chimeric porcine circovirus.
15. The vaccine as claimed in Claim 13 in the manufacture of a medicament for the
protection of a pig against viral infection or postweaning multisystemic wasting syndrome
(PMWS) caused by PCV2.

16. The vaccine as claimed in Claim 15, wherein the vaccine is the chimeric nucleic
acid molecule or the avirulent chimeric porcine circovirus in which said circovirus is a live,
avirulent chimeric porcine circovirus.
17. The vaccine as claimed in Claim 16 for parenteral, intranasal, intradermal or
transdermal administration.
18. The vaccine as claimed in Claim 17 for intralymphoid or intramuscular
administration.
19. A method of preparing the infectious chimeric nucleic acid molecule of PCV1-2 as
claimed in Claim 1, comprising removing an open reading frame (ORF) gene of a nucleic
acid molecule encoding an infectious, nonpathogenic PCV1; replacing the ORF gene
position of the PCV1 with an immunogenic ORF gene from a pathogenic PCV2; and
recovering the chimeric nucleic acid molecule.
20. The method as claimed in Claim 19, wherein said immunogenic PCV2 ORF gene
replaces the same ORF gene position of the PCVI nucleic acid molecule.
21. The method as claimed in Claim 20, wherein said immunogenic ORF gene is ORF2.
22. The method as claimed in Claim 21, wherein said ORF2 gene of PCV2 is obtained
from the molecular nucleic acid molecule of PCV2 contained in an expression vector having
ATCC Patent Deposit Designation PTA-3913.
23. An infectious PCV2 molecular DNA clone comprising ATCC Patent Deposit
Designation PTA-3913 or a PCV2 DNA clone derived therefrom.
24. A biologically functional plasmid or viral vector containing the infectious PCV2
molecular DNA clone as claimed in Claim 23.
25. An infectious reciprocal chimeric nucleic acid molecule of PCV2-1 comprising a
nucleic acid molecule encoding an infectious, pathogenic PCV2 which has an
immunogenic ORF2 gene from a nonpathogenic PCV1 in place of an ORF2 gene of the
PCV2 nucleic acid molecule.
The present invention relates to
infectious DNA clones, infectious chimeric DNA
clones of porcine circovirus (PCV), vaccines and
means of protecting pigs against viral infection or
postweaning multisystemic wasting syndrome (PMWS)
caused by PCV2). The new chimeric infectious
DNA clone and its derived, a virulent chimeric virus
are constructed from the nonpathogenic PCV1 in
which the immunogenic ORF gene of the pathogenic
PCV2 replaces a gene of the nonpathogenic PCV1,
preferably in the same position. The chimeric virus
advantageously retains the nonpathogenic phenotype
of PCV1 but elicits specific immune responses against
the pathogenic PCV2. The invention further embraces
the immunogenic polypeptide expression products.

Documents:

702-kolnp-2004-granted-abstract.pdf

702-kolnp-2004-granted-assignment.pdf

702-kolnp-2004-granted-claims.pdf

702-kolnp-2004-granted-correspondence.pdf

702-kolnp-2004-granted-description (complete).pdf

702-kolnp-2004-granted-drawings.pdf

702-kolnp-2004-granted-examination report.pdf

702-kolnp-2004-granted-form 1.pdf

702-kolnp-2004-granted-form 13.pdf

702-kolnp-2004-granted-form 18.pdf

702-kolnp-2004-granted-form 2.pdf

702-kolnp-2004-granted-form 3.pdf

702-kolnp-2004-granted-form 5.pdf

702-kolnp-2004-granted-gpa.pdf

702-kolnp-2004-granted-letter patent.pdf

702-kolnp-2004-granted-reply to examination report.pdf

702-kolnp-2004-granted-sequence listing.pdf

702-kolnp-2004-granted-specification.pdf


Patent Number 216040
Indian Patent Application Number 00702/KOLNP/2004
PG Journal Number 10/2008
Publication Date 07-Mar-2008
Grant Date 06-Mar-2008
Date of Filing 26-May-2004
Name of Patentee VIRGINIA TECH INTELLECTUAL PROPERTIES, INC. AND IOWA STATE UNIVERSITY RESEARCH FOUNDATION, INC.
Applicant Address 2200 KRFT DRIVE, SUITE 1050, BLACKSBURG, USA.
Inventors:
# Inventor's Name Inventor's Address
1 MENG XIANG-JIN- 401 CRAIG DRIVE BLACKS-DURG USA.
2 FENAUX MARTIJN 207 B, ARLINGTON ROAD REDWOOD CITY USA.
3 HALBUR PARTICK G 3211 KINGMANROAD AMES. USA.
PCT International Classification Number A61K
PCT International Application Number PCT/US02/39646
PCT International Filing date 2002-12-11
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 10/314, 512 2000-12-09 U.S.A.
2 60/340,775 2001-12-12 U.S.A.
3 60/424,840 2002-11-08 U.S.A.