Anda di halaman 1dari 6

Available online at www.sciencedirect.

com

Biologicals 37 (2009) 190e195


www.elsevier.com/locate/biologicals

Issues associated with residual cell-substrate DNA in viral vaccines


Li Sheng-Fowler, Andrew M. Lewis Jr., Keith Peden*
Division of Viral Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research,
Food and Drugs Administration, Bethesda, MD 20892, USA
Received 2 February 2009; accepted 2 February 2009

Abstract

The presence of some residual cellular DNA derived from the production-cell substrate in viral vaccines is inevitable. Whether this DNA
represents a safety concern, particularly if the cell substrate is derived from a tumor or is tumorigenic, is unknown. DNA has two biological
activities that need to be considered. First, DNA can be oncogenic; second, DNA can be infectious. As part of our studies to assess the risk of
residual cell-substrate DNA in viral vaccines, we have established assays that can quantify the biological activities of DNA. From data obtained
using these assays, we have estimated the risk of an oncogenic or an infectious event from DNA. Because these estimates were derived from the
most sensitive assays identified so far, they likely represent worst-case estimates. In addition, methods that inactivate the biological activities of
DNA can be assessed and estimations of risk reduction by these treatments can be made. In this paper, we discuss our approaches to address
potential safety issues associated with residual cellular DNA from neoplastic cell substrates in viral vaccines, summarize the development of
assays to quantify the oncogenic and infectivity activities of DNA, and discuss methods to reduce the biological activities of DNA.
Published by Elsevier Ltd on behalf of The International Association for Biologicals.

Keywords: Oncogenic DNA; Infectious DNA; Risk evaluation

1. Introduction: potential concerns associated with DNA chickens, such as H5N1 avian influenza viruses, has prompted
additional cell substrates to be evaluated for influenza vaccines,
The variety of cell substrates that have been used for the such as the Madin-Darby canine kidney cell line [3e5]. So far,
manufacture of viral vaccines licensed in the United States is many of the new mammalian cell substrates that are being
limited to primary cells of avian or monkey origin, to the diploid evaluated for viral vaccine manufacture are considered to be
cell lines (formerly termed diploid cell strains [1]) WI-38, MRC- neoplastic, since they have been immortalized by various
5, and FRhL-2, and to one continuous cell line, the VERO line mechanisms, and some are tumorigenic. The fear that compo-
(derived from African green monkey kidney cells) [2]. While nents derived from the production-cell substrate could induce
these cell substrates have produced vaccines of proven safety cancer in vaccine recipients was one of the reasons that the
and efficacy, it is increasingly apparent that this repertoire is Armed Forces Epidemiological Board recommended in 1954
insufficient for the production of the next generation of viral against the use of tumorigenic cells or cells derived from human
vaccines, such as those against HIV/AIDS, against emerging tumors for the manufacture of vaccines for human use [6,7].
infectious diseases (e.g., SARS), and against agents of bio- That this recommendation remained for over 40 years was due at
terrorism. In addition, the potential of a pandemic influenza least in part to the inability to evaluate this concern scientifically,
outbreak caused by influenza viruses that either cannot be which was due first to the inability to identify the risk factors and
propagated to high titers in eggs or that are pathogenic for second, once these risk factors were identified, to a lack of
assays capable of quantifying the risk posed by these factors.
* Corresponding author. Division of Viral Products, Center for Biologics It should be pointed out that this manuscript discusses potential
Evaluation and Research, Food and Drugs Administration, Building 29A,
Room 3D08, CBER, FDA, 29 Lincoln Drive, Bethesda, MD 20892, USA.
risks associated with use of novel highly tumorigenic neoplastic
Tel.: þ1 301 827 1708; fax: þ1 301 496 1810. cells for the manufacture of viral vaccines, and any discussion
E-mail address: keith.peden@fda.hhs.gov (K. Peden). about other types of cell substrates or products is beyond its scope.

1045-1056/09/$36.00 Published by Elsevier Ltd on behalf of The International Association for Biologicals.
doi:10.1016/j.biologicals.2009.02.015
L. Sheng-Fowler et al. / Biologicals 37 (2009) 190e195 191

2. Approaches to determine risk of cellular DNA cells were able to form tumors in vivo [13,16], one scientifically
based mechanism for the concern over the transfer of cancer-
Our general approach to risk assessment has been described inducing agents from vaccine to recipient was identified, viz.,
as a Defined Risks Approach (DRA) [8]. The algorithm that through DNA capable of expressing activated oncogenes.
CBER proposed for the DRA consists of five steps. 1. Iden- By analogy with viruses that integrate into the host
tifying the possible risk event; 2. Estimating or determining genome, a second mechanism for DNA-induced oncogenesis
the frequency with which the risk event might occur or has has been proposed, viz., through the integration of DNA. There
been observed to occur either in nature or under experimental are several ways that the integration of DNA could have
conditions; 3. Estimating the possible frequency of the risk oncogenic consequences. If DNA integrated next to a domi-
event per dose of vaccine; 4. Developing and determining the nant proto-oncogene such as c-myc and increased its expres-
sensitivity (with respect to the limits of the assay’s ability to sion level or activated its expression inappropriately, then this
detect the risk event) of one or more assays that can be use to could result in the oncogenic conversion of a normal cell to
detect the risk event; 5. Developing and validating one or more one with a neoplastic phenotype. If DNA integration occurred
processes that can be used to establish a product-specific in a tumor-suppressor gene, such as the p53 gene or the RB
safety factor to a defined level or to determine the level at gene, resulting in the functional inactivation of that gene, then
which current technology can be used to establish a safety this cell might, over time, become neoplastic through loss of
factor/limit. [Acceptable safety factors depend on the seri- heterozygosity by acquiring additional inactivating mutations
ousness of the risk event. In evaluating viral vaccines made in in the other allele. Both of these mechanisms have been seen
highly tumorigenic cells, the US FDA Vaccines and Related following retrovirus infection in both avian and mammalian
Biological Products Advisory Committee (VRBPAC) has systems. For example, activation of a proto-oncogene that
deemed a demonstrated risk of <1 in 107 to be acceptable with resulted in leukemia has been observed following infection of
respect to residual cellular DNA.] chickens with avian leukosis virus [17] and of mice with
Our approach to evaluate the risk of cell-substrate DNA Moloney murine leukemia virus [18,19], and the tumor-
from highly tumorigenic cells is as follows. 1. Develop suppressor gene p53 can be inactivated following retrovirus
sensitive and quantitative assays to measure the biological insertion [20,21].
activities of DNA. 2. Use data from the most sensitive assay to Clearly, therefore, integration of a retroviral proviral
estimate risks for a particular event; thus, estimates of risk genome can result in oncogenic events. The issue, though, is
would be conservative, since they should represent a worst- whether the frequency of integration of exogenous DNA is
case situation. 3. Use assays to quantify the amount of high enough to be of concern. Although this is not an easy
reduction in biological activity afforded by a particular treat- question to answer experimentally with cellular DNA due to
ment (e.g., nuclease digestion, chemical inactivation). 4. Use its sequence heterogeneity and genomic complexity, data
these data to estimate safety factors for a product with respect derived from studies with DNA vaccines, which have less
to the residual cell-substrate DNA in that product. complex genomes, suggest that integration of exogenous DNA
is an extremely low frequency event [22,23] and thus the
3. The biological activities of DNA frequency of integration at a particular site will be corre-
spondingly lower. Estimates have been made of the probability
DNA can have several measurable biological activities. of integration of DNA at a site that would result in the acti-
These include an oncogenic activity and an infectivity activity. vation of a cellular oncogene at approximately 1010 and for
Although not related to cell-substrate issues, DNA can also two independent events, as would be required to inactivate
have a transforming activity in bacterial and fungal systems. both alleles of a tumor-suppressor gene in a single cell, at
While the above DNA activities are mediated through products 1019 [24,25].
derived from gene expression (proteins, microRNA), another A second consideration for DNA integration is whether the
biological activity of DNA that does not require gene DNA from a tumorigenic cell would be any more of a concern
expression is the immunomodulatory activity of DNA itself than DNA of a normal diploid cell, since the differences in
[9,10]. While this immunomodulatory activity can be sequences between the two cell types are minimal, and inte-
measured in vivo, the amounts of DNA required for this effect gration with DNA is not site specific but occurs through ille-
are not typically present in vaccines. Therefore, based on gitimate recombination [26e31]. As such, the risk of DNA
current perceptions, the biological activities of DNA that need from an integration standpoint should be similar for all cellular
to be considered for vaccine safety are the oncogenic activity DNAs regardless of the phenotype of the cell from which it
and infectivity activity. was derived. Since amounts of DNA vaccines in the milligram
range have been approved for clinical evaluation, it is difficult
4. The oncogenic activity of DNA to imagine that the smaller quantities of residual cell-substrate
DNA present in viral vaccines would pose a significant risk
With the identification of oncogenes in the genomes of cells due to integration [32].
[11,12], the demonstration that certain activated cellular In conclusion, the major oncogenic concern with respect to
oncogenes were able to transform primary cells in vitro to DNA is considered to be via the introduction of a dominant
neoplastic cells [13e15], and that some of these transformed activated oncogene.
192 L. Sheng-Fowler et al. / Biologicals 37 (2009) 190e195

5. Infectivity activity of DNA facilitate a single cell taking up both oncogenes. In the second
approach, we evaluated both immune competent and immune
A second activity of DNA from neoplastic cell substrates incompetent mouse strains for their susceptibility to DNA-
that needs to be addressed as a potential safety issue is its induced tumor formation. We have identified a number of
infectivity activity [8,33]. This activity arises due to the strains that are more sensitive to oncogenic insult than the
potential presence of infectious virus genomes in the cellular newborn NIH Swiss mouse. At present, data indicate that
DNA either integrated or extrachromosomal. These genomes amounts of the ras/myc dual-expression plasmid down to 1 ng
could be those of DNA viruses or of the proviruses of retro- are capable of inducing tumors in mice (manuscript in prep-
viruses (exogenous or endogenous), since many viral genomes aration). At this level of DNA, the margin of safety from an
are infectious in vitro, such as polyomaviruses [34e37], oncogenic event from 10 ng of residual cell-substrate DNA is
papillomaviruses [38e42], adenoviruses [43e46], herpesvi- reduced from between 2.5  108 and 2.5  109 (estimated
ruses [47e52], parvoviruses [53e58], and retroviruses [59e from our earlier data [79]) to between 104 and 105. These
65], or in vivo, such as polyomaviruses [66], papillomaviruses estimations assume a haploid genome size of 3  109 base
[67,68], and retroviruses [69e75]. (The main concern with pairs for a mammalian genome and an oncogene size of
respect to retroviral DNA would be due to the presence of between 3000 and 30,000 base pairs. Although these revised
exogenous retroviruses, since all human cells have endoge- calculations are based on results with a plasmid that expresses
nous retroviruses and to date none has been shown to be both oncogenes, we feel that this is justified for two reasons.
infectious.) Therefore, the risk from the infectivity activity of First, our approach is to base estimates on the most sensitive
DNA would arise if the residual cell-substrate DNA encodes assay system. And second, we (unpublished results) and others
an infectious viral genome that, once inoculated, would [80] have found that a single oncogene can induce tumors in
produce an infectious virus in the transfected cell. If this virus mice. These lower estimates for the safety factor for an
were able to establish a productive infection in humans, the oncogenic event do not reach the 107 safety factors that have
consequences would be unpredictable. For these reasons, the been considered adequate by the US FDA Vaccines and
infectivity activity of DNA might represent more of a risk than Related Biological Products Advisory Committee (VRBPAC)
its oncogenic activity. Possibly the most obvious case where in 2005 with respect to residual DNA from tumorigenic cell
infectious cellular DNA would be a concern would be if an substrates (see transcripts at: www.fda.gov/ohrms/dockets/ac/
inactivated human immunodeficiency virus (HIV) vaccine 05/transcripts/2005-4188t1.pdf). In order to reach a safety
were produced from an infectious, replication-competent, factor of 107, methods to reduce the activity of DNA, such as
pathogenic HIV. While inactivation of the virus would be nuclease digestion and/or chemical inactivation, could be
monitored and the degree of inactivation quantified, the bio- incorporated into vaccine manufacture.
logical activity of the cellular DNA, which would contain the
infectious provirus, would also need to be addressed. 7. Development of assays to assess the infectivity activity
of DNA in vitro
6. Development of assays to assess the oncogenic activity
of DNA in vivo As stated above, should the vaccine cell substrate contain
the genome of an infectious virus either integrated or extra-
Because opinions on the risk posed by residual cellular chromosomal, this could be a safety issue, particularly for live
DNA in vaccines have varied from it representing no risk to it attenuated viral vaccines or inactivated viral vaccines manu-
being considered to be an important risk factor [76e78], and factured from pathogenic infectious viruses. To determine
because few studies had attempted to measure DNA oncoge- what levels of DNA represented an infectivity risk, we
nicity in vivo and thus few data were available, we undertook undertook studies to ascertain the specific infectivity in vitro
to investigate the oncogenic activity of cellular oncogenes in of the DNA of various viruses of different types. In our initial
an animal system with the aim of establishing a model that studies, we have determined the specific infectivity of the
could be used to estimate the risk of an oncogenic event by proviral DNA of the human immunodeficiency virus type 1
DNA. We generated expression plasmids for the human acti- (HIV-1) both as a plasmid infectious clone as well as the same
vated T24 H-ras gene and for the murine c-myc gene. Our virus integrated into the cellular genome. An in vitro trans-
initial studies demonstrated that these plasmids when injected fection/co-culture system was developed that could detect and
together were oncogenic in vivo, with NIH Swiss mice being quantify the infectivity of HIV-1 proviral DNA. This co-
more sensitive to oncogenic insult than C57BL/6, and newborn culture system can detect the infectivity of 1 pg of an HIV-1
mice more sensitive than adult mice. Both oncogenes were clone [81] and 2 mg of cellular DNA prepared from HIV-1-
required for tumor induction, and tumors were only induced at infected cells (Sheng-Fowler et al., manuscript submitted).
the highest amounts of DNA used (12.5 mg of each plasmid) From the specific infectivity of HIV-1 DNA, and knowing
[79]. Because this low efficiency of tumor induction would not the size of the diploid mammalian genome (6  109 base
permit the establishment of a practical assay, we sought ways pairs) and the HIV-1 genome (10,000 base pairs), we can
to increase this efficiency. In the first approach, we placed both estimate the amount of cellular DNA containing a single
oncogenes (human activated T24 H-ras gene and the murine c- provirus per cell that would correspond to 1 pg of viral DNA
myc gene) on the same molecule, reasoning that this should and produce an infection as: 1 pg O (104 O 6  109), which
L. Sheng-Fowler et al. / Biologicals 37 (2009) 190e195 193

equals 6  105 pg, or 600 ng. Thus, if the amount of residual the safety factors with respect to either DNA oncogenicity or
cell-substrate DNA in a product is 10 ng, then the safety factor DNA infectivity do not reach 107 when amounts of DNA
with respect to an infectious event for cellular DNA containing alone are considered, treatment of DNA with nucleases or
an infectious viral genome is 600 ng O 10 ng, or 60. If the cell chemicals, as appropriate for the vaccine, might be recom-
contains more than a single viral genome, then this safety mended when novel neoplastic cell substrates, such as highly
factor would be reduced accordingly. As stated above, safety tumorigenic cells or cells derived from human tumors, are to
factors of 107 have been considered appropriate with respect be used for viral vaccine manufacture.
to cell-substrate DNA, and thus, a safety factor of 60 or lower Finally, all decisions about the risks associated with any
would be insufficient. To obtain a safety factor in the 107 cell substrate need to be balanced both by the product type and
range, either the level of cell-substrate DNA would need to be the purity that the manufacturing process provides and the
lowered below 10 ng, or the biological activity of the DNA benefit of that product for the intended population.
would need to be reduced by nuclease digestion or chemical
inactivation. Assuming that only one copy of the retroviral Acknowledgements
DNA was present, then the amount of residual cell-substrate
DNA would need to be 10 fg or lower. However, if there were This work summarized in this paper was supported by
100 copies of the infectious viral genome, the amount of DNA the National Vaccine Program Office and a contract from the
would need to be reduced to 100 ag. Reducing residual cell- Division of Microbiology and Infectious Diseases of the
substrate DNA to these levels, even with the hardiest of viral National Institute of Allergy and Infectious Diseases through an
vaccines, would likely be impractical and difficult to docu- Interagency Agreement with CBER/FDA. We thank Phil
ment. Therefore, with certain cell substrates, additional treat- Krause, Hana Golding, Jerry Weir, Arifa Khan, and Robin Levis
ments of the DNA might be recommended. for comments on the manuscript.

8. Methods to reduce the biological activity of DNA


References
There are three general approaches to reduce the biological [1] Hayflick L. A brief history of cell substrates used for the preparation of
activity of DNA: nuclease digestion, chemical inactivation, human biologicals. Dev Biol 2001;106:5e23.
and irradiation. For live viral vaccines, nuclease digestion is [2] Plotkin S, Orenstein W, Offit P. Vaccines. 5th ed. Saunders Elsevier;
the only method that can be used, whereas, for inactivated 2008.
viral vaccines, subunit vaccines, or purified proteins, all three [3] Robertson JS, Nicolson C, Bootman JS, Major D, Robertson EW,
Wood JM. Sequence analysis of the haemagglutinin (HA) of influenza A
approaches have been used either separately or in combina- (H1N1) viruses present in clinical material and comparison with the HA
tion. As far as we are aware, there have been no studies that of laboratory-derived virus. J Gen Virol 1991;72:2671e7.
have quantified the amount of inactivation of a biological [4] Tree JA, Richardson C, Fooks AR, Clegg JC, Looby D. Comparison of
activity of DNA by these treatments. large-scale mammalian cell culture systems with egg culture for the
production of influenza virus A vaccine strains. Vaccine 2001;19:
Because of the sensitivity and dynamic range of the in vitro
3444e50.
infectivity assay, we have used this assay to quantify the [5] Medema JK, Meijer J, Kersten AJ, Horton R. Safety assessment of Madin
reduction in biological activity afforded by nuclease digestion Darby canine kidney cells as vaccine substrate. Dev Biol (Basel) 2006;
and by b-propiolactone (BPL) treatment. Results have shown 123:243e50 [discussion 265e6].
that either nuclease digestion or BPL treatment can reduce the [6] Hilleman MR. Cells, vaccines, and the pursuit of precedent. Natl Cancer
Inst Monogr 1968;29:463e9.
infectivity of DNA by more than 105 folds (Sheng-Fowler et al.,
[7] Hilleman MR. Line cell saga e an argument in favor of production of
submitted). Current work is directed at quantifying the reduction biologics in cancer cells. Adv Exp Med Biol 1979;118:47e58.
of DNA biological activity obtained by formaldehyde and [8] Lewis Jr AM, Krause P, Peden K. A defined-risks approach to the
binary ethylenimine, reagents also used to inactivate viral regulatory assessment of the use of neoplastic cells as substrates for viral
vaccines and that also inactivate the biological activity of DNA. vaccine manufacture. Dev Biol 2001;106:513e35.
[9] Ishii KJ, Gursel I, Gursel M, Klinman DM. Immunotherapeutic utility of
stimulatory and suppressive oligodeoxynucleotides. Curr Opin Mol Ther
9. Concluding remarks 2004;6:166e74.
[10] Rothenfusser S, Tuma E, Wagner M, Endres S, Hartmann G. Recent
Our studies on the oncogenicity and infectivity of DNA advances in immunostimulatory CpG oligonucleotides. Curr Opin Mol
have revealed that the levels at which both activities can be Ther 2003;5:98e106.
detected in sensitive in vivo or in vitro systems are lower than [11] Bishop JM, Baker B, Fujita D, McCombe P, Sheiness D, Smith K, et al.
Genesis of a virus-transforming gene. Natl Cancer Inst Monogr 1978;48:
hitherto demonstrated. Thus, both need to be considered with 219e23.
respect to the potential risks associated with residual cell- [12] Cooper GM. Cellular transforming genes. Science 1982;217:801e6.
substrate DNA. Because the oncogenicity and infectivity [13] Land H, Parada LF, Weinberg RA. Tumorigenic conversion of primary
activities were derived from sensitive assays, and because they embryo fibroblasts requires at least two cooperating oncogenes. Nature
do not take into account other factors such as that chromatin 1983;304:596e602.
[14] Santos E, Pulciani S, Barbacid M. Characterization of a human trans-
might be taken up and expressed less efficiently than naked forming gene isolated from T24 bladder carcinoma cells. Fed Proc 1984;
DNA, the estimated safety factors are conservative and most 43:2280e6.
likely represent worst-case scenarios. Nevertheless, because [15] Bos TJ. Oncogenes and cell growth. Adv Exp Med Biol 1992;321:45e99.
194 L. Sheng-Fowler et al. / Biologicals 37 (2009) 190e195

[16] Thompson TC, Southgate J, Kitchener G, Land H. Multistage carcino- [40] Brandsma JL, Xiao W. Infectious virus replication in papillomas induced
genesis induced by ras and myc oncogenes in a reconstituted organ. Cell by molecularly cloned cottontail rabbit papillomavirus DNA. J Virol
1989;56:917e30. 1993;67:567e71.
[17] Hayward WS, Neel BG, Astrin SM. Activation of a cellular onc gene by [41] McBride AA, Dlugosz A, Baker CC. Production of infectious bovine
promoter insertion in ALV-induced lymphoid leukosis. Nature 1981;290: papillomavirus from cloned viral DNA by using an organotypic
475e80. raft/xenograft technique. Proc Natl Acad Sci U S A 2000;97:5534e9.
[18] Tsichlis PN. Oncogenesis by Moloney murine leukemia virus. Anti- [42] Nasseri M, Meyers C, Wettstein FO. Genetic analysis of CRPV patho-
cancer Res 1987;7:171e80. genesis: the L1 open reading frame is dispensable for cellular trans-
[19] Tsichlis PN, Lazo PA. Virus-host interactions and the pathogenesis of formation but is required for papilloma formation. Virology 1989;170:
murine and human oncogenic retroviruses. Curr Top Microbiol Immunol 321e5.
1991;171:95e171. [43] Burnett JP, Harrington JA. Infectivity associated with simian adenovirus
[20] Ben David Y, Prideaux VR, Chow V, Benchimol S, Bernstein A. Inac- type SA7 DNA. Nature 1968;220:1245.
tivation of the p53 oncogene by internal deletion or retroviral integration [44] Burnett JP, Harrington JA. Simian adenovirus SA7 DNA: chemical,
in erythroleukemic cell lines induced by Friend leukemia virus. Onco- physical, and biological studies. Proc Natl Acad Sci U S A 1968;60:
gene 1988;3:179e85. 1023e9.
[21] Johnson P, Benchimol S. Friend virus induced murine erythroleukaemia: [45] Sutjipto S, Simmons DG. Turkey respiratory tract adenoviruses: in vitro
the p53 locus. Cancer Surv 1992;12:137e51. infectivity of purified DNA. Am J Vet Res 1981;42:495e7.
[22] Ledwith BJ, Manam S, Troilo PJ, Barnum AB, Pauley CJ, Griffiths TG, [46] Graham FL. Covalently closed circles of human adenovirus DNA are
et al. Plasmid DNA vaccines: investigation of integration into host infectious. Embo J 1984;3:2917e22.
cellular DNA following intramuscular injection in mice. Intervirology [47] Fleckenstein B, Bornkamm GW, Ludwig H. Repetitive sequences in
2000;43:258e72. complete and defective genomes of Herpesvirus saimiri. J Virol 1975;15:
[23] Ledwith BJ, Manam S, Troilo PJ, Barnum AB, Pauley CJ, Griffiths TG, 398e406.
et al. Plasmid DNA vaccines: assay for integration into host genomic [48] Allen GP, Randall CC. Biological properties of equine herpesvirus type 1
DNA. Dev Biol (Basel) 2000;104:33e43. DNA: transfectivity and transforming capacity. Infect Immun 1978;22:
[24] Temin HM. Overview of biological effects of addition of DNA molecules 34e40.
to cells. J Med Virol 1990;31:13e7. [49] Miller G, Grogan E, Heston L, Robinson J, Smith D. EpsteineBarr viral
[25] Kurth R. Risk potential of the chromosomal insertion of foreign DNA. DNA: infectivity for human placental cells. Science 1981;212:452e5.
Ann N Y Acad Sci 1995;772:140e51. [50] Cameron IR, Wilkie NM, Macnab JC. The infectivity of herpes simplex
[26] Coffin JM. Molecular mechanisms of nucleic acid integration. J Med virus DNA in rat embryo cells is enhanced synergistically by DMSO and
Virol 1990;31:43e9. glucose. J Virol Methods 1983;6:183e91.
[27] Bullock P, Forrester W, Botchan M. DNA sequence studies of simian [51] Tognon M, Cattozzo EM, Bianchi S, Romanelli MG. Enhancement of
virus 40 chromosomal excision and integration in rat cells. J Mol Biol HSVeDNA infectivity, in VERO and RS cells, by a modified calciume
1984;174:55e84. phosphate transfection technique. Virus Genes 1996;12:193e7.
[28] Mooibroek H, Arnberg AC, de Jong B, Venema G. Effect of concen- [52] Kaaden OR. Transfection studies in vitro and in vivo with isolated
tration on the subsequent fate of plasmid DNA in human fibroblasts. Mol Marek’s disease virus DNA. IARC Sci Publ 1978;24:627e33.
Gen Genet 1985;199:82e8. [53] Casal JI, Diaz-Aroca E, Ranz AI, Manclus JJ. Construction of an
[29] Ruley HE, Fried M. Clustered illegitimate recombination events in infectious genomic clone of porcine parvovirus: effect of the 50 -end on
mammalian cells involving very short sequence homologies. Nature DNA replication. Virology 1990;177:764e7.
1983;304:181e4. [54] Laughlin CA, Tratschin JD, Coon H, Carter BJ. Cloning of infectious
[30] Stary A, Sarasin A. Molecular analysis of DNA junctions produced by adeno-associated virus genomes in bacterial plasmids. Gene 1983;23:
illegitimate recombination in human cells. Nucleic Acids Res 1992;20: 65e73.
4269e74. [55] Merchlinsky MJ, Tattersall PJ, Leary JJ, Cotmore SF, Gardiner EM,
[31] Yarom R, Lapidot A, Neer A, Baran N, Manor H. ‘Illegitimate’ Ward DC. Construction of an infectious molecular clone of the autono-
recombination events in polyoma-transformed rat cells. Gene 1987;59: mous parvovirus minute virus of mice. J Virol 1983;47:227e32.
87e98. [56] Senapathy P, Carter BJ. Molecular cloning of adeno-associated virus
[32] Griffiths E. Major issues associated with the use of cell substrates for the variant genomes and generation of infectious virus by recombination in
production of vaccines. Dev Biol 2001;106:25e35. mammalian cells. J Biol Chem 1984;259:4661e6.
[33] Krause PR, Lewis Jr AM. Safety of viral DNA in biological products. [57] Zhi N, Zadori Z, Brown KE, Tijssen P. Construction and sequencing of
Biologicals 1998;26:317e20. an infectious clone of the human parvovirus B19. Virology 2004;318:
[34] Diderholm H, Wesslen T. Infection of naturally insusceptible cells with 142e52.
DNA of polyoma and SV40 viruses. Acta Pathol Microbiol Scand 1963; [58] Samulski RJ, Berns KI, Tan M, Muzyczka N. Cloning of adeno-associ-
59:271e2. ated virus into pBR322: rescue of intact virus from the recombinant
[35] Di Mayorca GA, Eddy BE, Stewart SE, Hunter WS, Friend C, Bendich A. plasmid in human cells. Proc Natl Acad Sci U S A 1982;79:2077e81.
Isolation of infectious deoxyribonucleic acid from SE polyoma-infected [59] Fisher AG, Collalti E, Ratner L, Gallo RC, Wong-Staal F. A molecular
tissue cultures. Proc Natl Acad Sci U S A 1959;45:1805e8. clone of HTLV-III with biological activity. Nature 1985;316:262e5.
[36] Orth G, Atanasiu P, Boiron M, Rebiere JP, Paoletti C. Infectious and [60] Adachi A, Gendelman HE, Koenig S, Folks T, Wiley R, Rabson A, et al.
oncogenic effect of DNA extracted from cells infected with polyoma Production of acquired immunodeficiency syndrome-associated retro-
virus. Proc Soc Exp Biol Med 1964;115:1090e5. virus in human and nonhuman cells transfected with an infectious
[37] Gerber P. An infectious deoxyribonucleic acid derived from vacuolating molecular clone. J Virol 1986;59:284e91.
virus (SV40). Virology 1962;16:96e7. [61] Barker CS, Pickel J, Tainsky M, Hunter E. Molecular cloning of the
[38] Watts SL, Ostrow RS, Phelps WC, Prince JT, Faras AJ. Free cottontail MasonePfizer monkey virus genome: biological characterization of
rabbit papillomavirus DNA persists in warts and carcinomas of infected genome length clones and molecular comparisons to other retroviruses.
rabbits and in cells in culture transformed with virus or viral DNA. Virology 1986;153:201e14.
Virology 1983;125:127e38. [62] Itohara S, Sekikawa K. Molecular cloning of infectious proviral genomes
[39] Brandsma JL, Yang ZH, Barthold SW, Johnson EA. Use of a rapid, of bovine leukemia virus. Virology 1987;159:158e60.
efficient inoculation method to induce papillomas by cottontail rabbit [63] Olmsted RA, Barnes AK, Yamamoto JK, Hirsch VM, Purcell RH,
papillomavirus DNA shows that the E7 gene is required. Proc Natl Acad Johnson PR. Molecular cloning of feline immunodeficiency virus. Proc
Sci U S A 1991;88:4816e20. Natl Acad Sci U S A 1989;86:2448e52.
L. Sheng-Fowler et al. / Biologicals 37 (2009) 190e195 195

[64] Regier DA, Desrosiers RC. The complete nucleotide sequence of inoculation of plasmid DNA encoding full-length SIVmac239. AIDS Res
a pathogenic molecular clone of simian immunodeficiency virus. AIDS Hum Retroviruses 1999;15:445e50.
Res Hum Retroviruses 1990;6:1221e31. [73] Kent SJ, Dale CJ, Preiss S, Purcell DF. Evidence of recombination
[65] Inabe K, Ikuta K, Aida Y. Transmission and propagation in cell culture of between 30 and 50 LTRs in macaques inoculated with SIV DNA. AIDS
virus produced by cells transfected with an infectious molecular clone of Res Hum Retroviruses 2002;18:227e30.
bovine leukemia virus. Virology 1998;245:53e64. [74] Kent SJ, Dale CJ, Preiss S, Mills J, Campagna D, Purcell DF. Vaccination
[66] Israel MA, Chan HW, Hourihan SL, Rowe WP, Martin MA. Biological with attenuated simian immunodeficiency virus by DNA inoculation.
activity of polyoma viral DNA in mice and hamsters. J Virol 1979;29: J Virol 2001;75:11930e4.
990e6. [75] Purcell DF, Cameron PU, Mills J, Kent S. Infectivity of wild-type and
[67] Brandsma JL, Yang ZH, DiMaio D, Barthold SW, Johnson E, Xiao W. deleted proviral SIV DNA. Dev Biol 2001;106:395e406.
The putative E5 open reading frame of cottontail rabbit papillomavirus is [76] Petricciani JC, Horaud FN. DNA, dragons and sanity. Biologicals 1995;
dispensable for papilloma formation in domestic rabbits. J Virol 1992;66: 23:233e8.
6204e7. [77] Petricciani JC, Regan PJ. Risk of neoplastic transformation from cellular
[68] Kreider JW, Cladel NM, Patrick SD, Welsh PA, DiAngelo SL, DNA: calculations using the oncogene model. Dev Biol Scand 1987;68:
Bower JM, et al. High efficiency induction of papillomas in vivo using 43e9.
recombinant cottontail rabbit papillomavirus DNA. J Virol Methods [78] Petricciani J, Loewer J. An overview of cell DNA issues. Dev Biol 2001;
1995;55:233e44. 106:275e82.
[69] Letvin NL, Lord CI, King NW, Wyand MS, Myrick KV, Haseltine WA. [79] Sheng L, Cai F, Zhu Y, Pal A, Athanasiou M, Orrison B, et al. Onco-
Risks of handling HIV. Nature 1991;349:573. genicity of DNA in vivo: tumor induction with expression plasmids for
[70] Willems L, Portetelle D, Kerkhofs P, Chen G, Burny A, Mammerickx M, activated H-ras and c-myc. Biologicals 2008;36:184e97.
et al. In vivo transfection of bovine leukemia provirus into sheep. [80] Burns PA, Jack A, Neilson F, Haddow S, Balmain A. Transformation of
Virology 1992;189:775e7. mouse skin endothelial cells in vivo by direct application of plasmid
[71] Willems L, Thienpont E, Kerkhofs P, Burry A, Mammerickx M, DNA encoding the human T24 H-ras oncogene. Oncogene 1991;6:
Kettmann R. In vivo infection of sheep by bovine leukemia virus 1973e8.
mutants. J Virol 1993;67:4078e85. [81] Peden K, Sheng L, Pal A, Lewis A. Biological activity of residual
[72] Liska V, Khimani AH, Hofmann-Lehmann R, Fink AN, Vlasak J, cell-substrate DNA. Dev Biol (Basel) 2006;123:45e53 [discussion
Ruprecht RM. Viremia and AIDS in rhesus macaques after intramuscular 55e73].

Anda mungkin juga menyukai