Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Nov 5.
Published in final edited form as: Vaccine. 2009 Nov 5;27S4:D73–D79. doi: 10.1016/j.vaccine.2009.07.103

Smallpox Vaccines for Biodefense

Richard B Kennedy 1,2,3, Inna Ovsyannikova 1,2, Gregory A Poland 1,2,3,*
PMCID: PMC2764553  NIHMSID: NIHMS136803  PMID: 19837292

Abstract

Few diseases can match the enormous impact that smallpox has had on mankind. Its influence can be seen in the earliest recorded histories of ancient civilizations in Egypt and Mesopotamia. With fatality rates up to 30%, smallpox left its survivors with extensive scarring and other serious sequelae. It is estimated that smallpox killed 500 million people in the 19th and 20th centuries. Given the ongoing concerns regarding the use of variola as a biological weapon, this review will focus on the licensed vaccines as well as current research into next-generation vaccines to protect against smallpox and other poxviruses.

Keywords: smallpox vaccine, variola virus, vaccinia virus, biodefense

History

Smallpox is thought to have originated near Egypt and the Near East between 5,000 and 10,000 years ago. [1] From here the disease spread into Asia, Europe and northern Africa. Smallpox was brought to the western hemisphere by European explorers and military expeditions with devastating results. It is estimated that it killed millions and contributed to the downfall of the Incan and Aztec empires. [2] By the 17th century, smallpox was endemic throughout most of the world. By the early 1900’s a far milder version of smallpox (alastrim - caused by variola minor) began to circulate throughout North and Central America and in the UK.

In 1796 Edward Jenner published his landmark findings that vaccination with cowpox would prevent infection with smallpox. [3, 4] While the concept of vaccination spread rapidly throughout the world, it was not until the Intensified Smallpox Eradication Program of 1967 that much progress was made in controlling the disease on a large scale. [2] Massive vaccination campaigns occurred worldwide and were combined with sophisticated surveillance systems to detect outbreaks, which were then contained using the ring vaccination approach. [5] The last naturally occurring case of smallpox was in 1977, and one year later smallpox claimed its last human life after a laboratory exposure in England. In May of 1980 the World Health Organization declared that smallpox had been eradicated. [6]

Pathology

Infection typically occurred primarily through the respiratory route or contact with contaminated clothing and bedding, although airborne exposure cannot be ruled out.[2, 7] This was followed by a prolonged, asymptomatic incubation period of 12-14 days which culminated in a 2-4 day prodromal phase marked by high fever, headache, backache, nausea and prostration. [8, 9] The characteristic rash began on the face and extremities and eventually covered the entire body. The lesions begin as small reddish macules which, over the next several weeks, enlarge and become pustules several millimeters in size. The lesions eventually scab over leaving behind extensive scarring. Other long-term sequelae can include blindness, encephalitis, secondary infections and arthritis. Clinical symptoms varied widely and are likely attributable to infectious dose, host health, pre-existing immunity and genetic factors of both host and virus. Fatality rates can be as high as 30%, with death usually attributed to toxemia, hypotension and multi-organ failure; however the exact causes of death are not fully understood and it is likely that both viral cytopathic effects and inflammatory immune mediators contribute to mortality. [9]

Prevention and Treatment

The earliest effective preventive measure for smallpox was variolation, the intentional introduction of smallpox material into a healthy host. [1, 2] There were two main techniques: 1) dried scab material inhalation, and 2) pus from active lesions scratched into the skin. Both approaches resulted in full-fledged cases of smallpox, the vast majority of which had far milder symptoms with case fatality rates of 1-2%. One of the first large-scale uses of this treatment in North America was when General Washington ordered the variolation of US troops during the Revolutionary War. [10]

The eradication efforts were made possible, in part, by the availability of an effective vaccine. Although Jenner’s initial work focused on cowpox, by the mid twentieth century most vaccines were based on vaccinia, an immunologically cross-protective poxvirus. Although the origins of vaccinia virus are unknown, the vaccine was remarkably effective, with take rates over 97%. Protection was believed to last for 3-10 years and revaccination was required every 10 years. [2] Recent research has shown that immune responses to vaccinia can be detected over 70 years after vaccination, however, the level of protection afforded by these immune markers is not known. [11-18]

There are few treatments for smallpox, underscoring the need for safe and effective vaccines. Hyperimmune serum obtained from recent vaccine recipients has been used to produce vaccinia immune globulin (VIG) which is used to treat both smallpox outbreaks and vaccine complications. [19] Similarly, cidofovir and related anti-viral agents have shown inhibitory activity against poxviruses in vitro and in animal challenge models. [20-22] Poxvirus antivirals are an area of active research and are likely to play key roles supplementing vaccines in biodefense preparations against smallpox.

Smallpox as a bioweapon

With the recent rise in terrorism activity (including bioterrorism), and concerns regarding state-sponsored biological weapons programs have led to significant efforts in the areas of biodefense and biopreparedness. [23] Both the Soviet Union and Iraq are known to have had large biological weapons programs which included smallpox. [24, 25] Variola major, the causative agent of smallpox is considered a top bioterrorism agent. [26] 1) It is a disfiguring illness with a high mortality rate, 2) it is a highly contagious pathogen with an extremely low infectious dose, 3) the disease has a long, asymptomatic incubation period, 4) there are no effective treatments and limited vaccine stocks, 5) an outbreak would cause widespread panic and societal disruption and, 6) since vaccination ceased over 30 years ago, and the present population is largely susceptible and vaccine recipients are likely to have waning immunity. Notably a contemporary release of smallpox would not be its first use as a weapon (Table 1).

Table 1. The use of Smallpox as a Bioweapon.

Year Incident
1500’s Spanish explorers bring smallpox-contaminated items to American Indians.
Resulting outbreaks likely kill millions.
1760’s French and Indian War - British army gives contaminated blankets to
Indian tribesallied with the French, with mortality rates reaching 50%.
1776 Revolutionary war - British forces in Boston send infected civilians out to
Continental Army, causing outbreaks among American soldiers.
1800’s Westward spread of American settlers - Contaminated items given to
Indian tribes causing wide-spread outbreaks.
1860’s Civil War - Contaminated clothing sold to Union troops
1980’s - ? Numerous countries, including Russia, Iraq, Syria, North Korea, and China
have robust biological weapons programs.

Current Vaccines

First generation vaccines developed during the eradication effort consist of live vaccinia virus (See Table 2). Dryvax®, the vaccine used in the US is based on the New York City Board of Health (NYCBOH) strain of vaccinia and together with the Lister strain used in Europe, Africa and Asia was deemed safer than other vaccine strains. [2] First generation vaccines were produced by infecting the abdomen and flanks of calves or other large animals. The infected areas were then scraped to collect the lymph exudate and virus was purified, stabilized with glycerol and phenol, and in many cases lyophilized. The resulting vaccine was introduced into the epidermal layer of the skin using a bifurcated needle with multiple punctures. Production of these vaccines ceased in the 1970’s and 1980’s.

Table 2. Smallpox vaccines and vaccine candidates.

Vaccine Name VACV Strain
Details
Conventional Vaccines
Dryvax NYCBOH Lyophilized vaccine used in US during eradication.[27]
APSV NYCBOH Frozen liquid vaccine used in US during eradication.[34]
Lancy-Vaxina Lister Lyophilized vaccine used world-wide during eradication.[27]
Tissue culture based, live virus vaccines
ACAM1000 NYCBOH Grown in MRC-5 cells. Improved safety profile in animals,
immunogenicity similar to Dryvax.[45]
ACAM2000 NYCBOH Grown in Vero cells Improved safety profile in animals,
immunogenicity similar to Dryvax.[45]
CCSV NYCBOH Tissue culture vaccine grown in MRC-5 cells. Similar
immunogenicity to Dryvax.[57]
Elstree-BN Lister Immunogenicity is equivalent to conventional Lister vaccines and
Dryvax.[51]
Replication competent, attenuated vaccines
MVA Ankara Passaged through chick embryo fibroblasts. Lost 15% of genome
and cannot replicate in human cells. Used at end of eradication with
few adverse events.[62]
LC16m8 Lister Attenuated vaccine used in Japan at end of eradication era with an
improved safety profile.[59]
NYVAC Copenhagen Attenuated strain missing 18 ORFs. Improved safety profile but has
not been widely used.[67]
dVV-L Lister UDG gene has been deleted to improve safety but has no not been
widely used.[72]
IMVAMUNE MVA Bavarian Nordic reformulation of MVA vaccine.[65]
TBC-MVA MVA Therion Biologics reformulation of MVA vaccine.[66]
Subunit vaccines
Protein VACV H3L based[98]; A33R, B5R, L1R, based[103]; Polyvalent w/ D8
protein.[127]
DNA VACV A27L, A33R, L1R, B5R based[105]; A27L, A33R and B5R
based.[128]
DNA VARV A30, B7, F8 proteins.[109]

Three to four days following successful vaccination a pock forms at the vaccine site as a result of local viral replication and inflammatory response. Over the first week, the pock grows to 10-15 mm in size, fills with pus and eventually drains and scabs over. Approximately 3-4 weeks after vaccination the scab will fall off leaving a small scar.

Humoral responses begin to develop within 2 weeks of vaccination, with neutralizing antibody titers peaking around day 28. [16, 27] Antibody titers decline in the first several years following vaccination and then stabilize and remain constant for decades. [11] Robust CD4 and CD8 T cell responses occur after immunization and reach their peak between 2-5 weeks post vaccination. Similar to humoral immunity, virus-specific T cells can be detected for decades following vaccination, with CD4+ T cells showing greater longevity. [11] Unfortunately, the magnitude of the protective immune response has not been clearly defined. One study showed that subjects with neutralizing antibody titers > 1:32 were less susceptible to infection[28], while a similar study found that titers >1:20 were protective. [29] Unfortunately, no similar data are available for cell-mediated responses.

The live virus vaccines used during the eradication efforts were not without side-effects and complications, in fact they are among the most reactogenic of all FDA-approved vaccines. Common side effects include: fever, headache, pruritis, lymphadenopathy, fatigue, muscle aches and nausea. Although rare, life-threatening conditions such as postvaccinal encephalitis, progressive vaccinia and eczema vaccinatum, myopericarditis or generalized vaccinia can occur. [30-32] Historically, for every million vaccine recipients there were 1-2 deaths and hundreds of cases with complications severe enough to require hospitalization. [33] If the current population of the United States were vaccinated, it might result in several hundred deaths and tens of thousands of hospitalizations. In the absence of endemic smallpox this risk was not acceptable, and large-scale vaccination halted in 1972 in the United States and by 1980 in most other countries as well. From a biodefense standpoint these vaccine safety concerns would be of secondary importance to the need to quickly and efficiently break the chain of transmission and contain the outbreak.

As concerns regarding bioterrorism increased in the earliest years of the 21st century, national stockpiles of smallpox vaccines were found to be extremely limited. In 2005 the stockpile was estimated to have 15 million doses of lyophilized Dryvax vaccine and 85 million doses of frozen Aventis Pasteur Smallpox Vaccine (APSV). [34] Other countries were found to have similarly limited stockpiles of smallpox vaccine, enough for an estimated 10% of the world population. [35] In the early 2000’s the US government re-instituted large scale vaccination programs among the military and among civilian healthcare workers. [32, 36, 37] Consequently considerable effort was put into extending the current stocks and development of replacement vaccines. Given the lack of endemic smallpox all new smallpox vaccines are being compared to Dryvax® in terms of both safety and immunogenicity. [38] Controlled studies indicated that Dryvax and other first-generation vaccines (APSV, Lister) could be diluted at least 5-fold without a significant drop in either take rates or immunogenicity[39-43], although other groups have argued that dilution will increase the risk of ineffective inoculations and may hamper ring vaccine strategies. [44]

Acambis and Baxter laboratories plaque purified a NYCBOH isolate from Dryvax® with an improved safety profile in animal models and used this strain to create a cell-culture based vaccine; ACAM2000. [45, 46] This vaccine has been through multiple clinical trials and was found to have the same take rate, lesion size and a similar rate of adverse events as Dryvax. [47] Neutralizing antibody titers induced by ACAM2000 were slightly lower than Dryvax in one study[47] but equivalent in others[45, 48], while T cell responses showed no significant differences. In a non-human primate study, ACAM2000 was found to be as effective as Dryvax in preventing disease following a lethal monkeypox challenge. [49] Interestingly, ACAM2000 takes rates decreased significantly upon dilution. [48] ACAM2000 received FDA approval in 2007[50]. By 2004 Acambis had produced 200 million doses of the vaccine for the national stockpile[45] and is contracted to produce more.

Development of New Vaccines

In addition to licensed vaccines, a number of other vaccine candidates are in various stages of development. [51-54] These include other live vaccinia strains, live attenuated strains deficient in one or more viral genes (believed to be safer alternatives to conventional vaccinia strains), and inactivated or subunit vaccines that potentially offer even greater safety profiles.

Live smallpox vaccines produced in tissue culture

Initial work on tissue culture smallpox vaccines was performed in the 1970’s, and again emphasized in the early 2000’s as replacements for the aging calf-lymph products. These vaccines sought to retain the immunogenicity of the live vaccines while controlling the production conditions in order to curtail non-sterile handling of the vaccine components, and eliminate bacterial, viral, and fungal adventitious agents. [55] These vaccine candidates include: ACAM1000 and ACAM2000 produced by Acambis from plaque purified NYCBOH isolates grown in MRC-5 and Vero cells respectively[45, 56], Cell-culture smallpox vaccine (CCSV), another NYCBOH-based vaccine grown in MRC-5 cells and developed by DynPort, [57], and Elstree-BN, a Bavarian-nordic product based on the Lister strain.[51] Most of these strains have undergone extensive side-by-side testing with Dryvax® or Lister vaccines and have similar safety and immunogenicity profiles. [58]

Replication competent, attenuated smallpox vaccines

During the eradication effort a number of attempts were made to create less pathogenic virus strains with a corresponding increase in vaccine safety. [34] While most of these newer strains were not sufficiently immunogenic for use in vaccines, several promising candidates remain.

LC16m8 was developed through serial passage of VACV Lister through rabbit kidney cells. [59, 60] The resulting strain was less virulent and was used in Japan in the 1970’s. [60] LC16m8 was found to have similar take rates and levels of neutralizing antibodies than Lister, with fewer adverse events and febrile reactions. [61] LC16m8 is one of the candidates for the creation of a US stockpile of attenuated smallpox vaccine.

Modified Virus Ankara (MVA) was developed through the serial passage of VACV Ankara through chick embryo fibroblasts. [62] After 572 passages, the virus lost ~15% of its genome and the ability to replicate in most mammalian cells. [63] It was used in Germany in the late 1960’s to vaccinate over 100,000 individuals with very few adverse events. As smallpox did not circulate in Germany during or after this time period, there are no efficacy data on MVA. Newer versions of MVA are being produced by a number of manufacturers including: ACAM3000 by Acambis (www.clinicaltrials.gov # NCT00079820 last accessed 7/14/2009), IMVAMUNE by Bavarian Nordic[64, 65], and TBC-MVA by Therion Biologics Corporation[66]. MVA is also a top candidate for inclusion in the US stockpile as an attenuated vaccine.

NYVAC, another highly attenuated vaccine strain was derived from the parental Copenhagen vaccinia strain by deletion of 19 ORFs including the thymidine kinase, ribonucleotide reductase, complement binding, hemagglutinin and other genes.[67] While NYVAC is deemed to be a “safer” vaccine candidate, direct comparisons with Dryvax or Lister-based vaccines have shown that NYVAC possesses inferior immunogenicity. [18, 68, 69] From the standpoint of biodefense preparations against poxviruses, NYVAC is not an optimal vaccine candidate. On the other hand, foreign antigens can be readily incorporated in NYVAC based vectors and induce robust immunity. NYVAC is therefore a promising vehicle for the development of vaccines against other diseases. [69-71]

A replication defective vaccine based on the Lister strain (dVV-L) was created through the deletion of the uracil-DNA-glycosylase (UDG) gene. [72] The resulting vaccine, while replication incompetent outside of certain cell lines which provide exogenous UDG is still capable of initial infection and early gene expression. Early work found that this vaccine candidate had an enhanced safety profile compared to wild-type Lister vaccine. [73] Mouse vaccination and lethal challenge studies found that dVV-L induced protective T and B cell responses against cowpox or vaccinia virus but required higher vaccine doses to prevent death in response to more virulent natural mouse pathogen, ectromelia virus. [74]

The Dairen strain was used primarily in Japan during the eradication effort and more recently was used to create the attenuated Dairen-I strain.[75, 76] This strain lacks several host range and immunomodulatory genes [77] which restrict its replicative potential and may enhance its safety profile. Similar to NYVAC, Dairen-I has served as a useful vector in the development of vaccines against mycobacterium, HIV, SARS and other pathogens. [78-80]

A variety of other attenuated vaccinia virus strains have been created through the selected deletion or alteration of host range genes, virulence factors and immunomodulatory genes. [81-87] These recombinant vaccinia strains have been shown to grow to lower titers in various organs, and/or exhibit less toxicity and morbidity in immunocompromised animal models. Other groups have used genetic engineering to incorporate growth factors, cytokines, chemokines and innate immune activators into vaccinia virus in an attempt to boost immune responses after infection. [88]

Overall, attenuated vaccinia strains are far safer in animal models, especially those with defective immune responses, and in a number of cases (MVA, LC16m8) induce fewer side effects in human populations. Unfortunately a growing body of literature indicates that they are less immunogenic than their conventional vaccine counterparts, requiring high doses of antigen and/or repeat immunization to induce similar levels of antibody titers or cellular immunity.[18] Another interesting observation is that subclones with markedly different behavioral characteristics exist within live viral stocks, and outgrowth of these lines may have unforeseen effects on safety, immunogenicity or both.[89] In the absence of endemic smallpox it is not known if these vaccines will provide full protection or merely attenuate disease symptoms. One potential option would be to use these safer vaccines in individuals with compromised immunity, while another approach would be to provide these vaccines to everyone in the population as a priming dose. In this case, a partially protected population may serve as a deterrent to bioterrorism and the conventional vaccine would always be available in the case of an outbreak or biological attack. In any case, these attenuated vaccine formulations are a valuable component of biodefense preparations.

Inactivated smallpox vaccines

In the mid-20th century a great deal of effort went into the development of inactivated smallpox vaccines, with limited success. [90, 91] Although these vaccines exhibited far greater safety profiles, and induced strong humoral immunity; the resulting responses were unable to control infection and prevent illness in animal models. [92, 93] This may be attributed to the lack of key immunogenic epitopes within the killed vaccines. These formulations lack the extracellular enveloped virions which contain an extra lipid membrane with several additional viral proteins. [94-96] These proteins are targets of neutralizing antibody responses and immunity to both the intracellular and extracellular virions is likely needed for complete protection.

Subunit-based smallpox vaccines

The development of protein, peptide or nucleic acid based subunit vaccines for preventing smallpox is an active area of investigation. Most groups have focused on the inclusion of viral proteins targeted by humoral immunity, although with the recent identification of CD4 and CD8 T cell epitopes, cellular responses to poxviruses are beginning to be taken into account as well. [97, 98] [99] Several groups have found that combinations of membrane and virion proteins (A27L, A33R, B5R, H3L, L1R) or DNA are far more effective than single protein vaccines and provide excellent protectin in both mice and non-human primate models.[99-106] One consideration that must be taken into account is the genetic diversity of poxviruses. The vaccines are all based on vaccinia virus whereas the most important pathogenic poxviruses are variola and monkeypox. Cross-protective immunity is a direct result of antigenic similarity between the viruses. Using whole viruses provides a large mixture of vaccinia antigens, many of which are highly conserved between poxvirus strains. With protein or DNA based subunit vaccines the selection of antigens is severely limited. Heterogeneity within individual proteins may have negative consequences on vaccine efficacy. [107, 108] One alternative to this is to base vaccine components on variola homologs rather than the vaccinia proteins. [109] The identification of HLA class I and class II restricted epitopes from poxviruses has opened the door to the development of safe and immunogenic peptide-based vaccines. [110-117] [118]

Another area of investigation is the use of different delivery routes, including mucosal vaccines [119, 120] and plant-based formulations. [121]

Conclusions and looking to the future

Given the tremendous effect a biological attack with smallpox would cause, it is imperative that we are properly prepared. Vaccines are among the most cost-effective of healthcare measures and play key roles in biodefense preparations both through deterrent action and as preventive measures. Although several elements of our bio-preparedness efforts are now being accomplished (sufficient vaccine doses for the US population, new anti-virals), much work remains as many countries have insufficient doses for their population and live virus vaccines remain problematic in today’s environment of extreme sensitivity to adverse vaccine reactions. Numerous potentially safer vaccine products are in various stages of research and development but will require rigorous testing to ensure that they induce immunity equivalent to conventional smallpox vaccines. To this end, further work needs to be done to refine animal models for testing new smallpox vaccines as small animal studies using different poxviruses can produce conflicting results, and non-human primate models do not fully recapitulate human exposure to smallpox. [122]

Another avenue of research that should be further developed is that of immunogenetics and understanding why some individuals are predisposed to ineffective vaccine responses or even adverse events. [123-125] The development of simple screening tests would allow us to steer individual patients toward vaccines best suited to their individual requirements. [126]

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Dixon CW. Smallpox. J. & A. Churchill; London: 1962. [Google Scholar]
  • [2].Fenner F, Arita I, Jezek Z, Ladnyi ID. Smallpox and its Eradication. W.H.O.; Geneva: 1988. DAH. [Google Scholar]
  • [3].Jenner E. An inquiry into the causes and effects of the variolae vaccinae, a disease discovered in some of the western counties of England, particularly Gloucestershire, and known by the name of the cow pox. Law; London: p. 1798. [Google Scholar]
  • [4].Cook GC. The smallpox saga and the origin(s) of vaccination. Journal of the Royal Society of Health. 1996 Aug;116(4):253–5. doi: 10.1177/146642409611600412. [DOI] [PubMed] [Google Scholar]
  • [5].Fenner F. A successful eradication campaign. Global eradication of smallpox. Rev Infect Dis. 1982 Sep-Oct;4(5):916–30. doi: 10.1093/clinids/4.5.916. [DOI] [PubMed] [Google Scholar]
  • [6].Fenner F. The global eradication of smallpox. Med J Aust. 1980 May 17;1(10):455–5. doi: 10.5694/j.1326-5377.1980.tb135034.x. [DOI] [PubMed] [Google Scholar]
  • [7].Wehrle PF, Posch J, Richter KH, Henderson DA. An airborne outbreak of smallpox in a German hospital and its significance with respect to other recent outbreaks in Europe. Bull World Health Organ. 1970;43(5):669–79. [PMC free article] [PubMed] [Google Scholar]
  • [8].Neff JM, Lane JM, Fulginiti VA. Smallpox and smallpox vaccination. N Engl J Med. 2003 May 8;348(19):1920–5. author reply -5. [PubMed] [Google Scholar]
  • [9].Moore ZS, Seward JF, Lane JM. Smallpox. Lancet. 2006 Feb 4;367(9508):425–35. doi: 10.1016/S0140-6736(06)68143-9. [DOI] [PubMed] [Google Scholar]
  • [10].Fenn EA. Pox Americana: the great smallpox epidemic of 1775-82. 1st ed. Hill and Wang; New York: 2001. [PubMed] [Google Scholar]
  • [11].Hammarlund E, Lewis MW, Hansen SG, Strelow LI, Nelson JA, Sexton GJ, et al. Duration of antiviral immunity after smallpox vaccination. Nat Med. 2003 Sep;9(9):1131–7. doi: 10.1038/nm917. [DOI] [PubMed] [Google Scholar]
  • [12].Crotty S, Felgner P, Davies H, Glidewell J, Villarreal L, Ahmed R. Cutting edge: long-term B cell memory in humans after smallpox vaccination. J Immunol. 2003 Nov 15;171(10):4969–73. doi: 10.4049/jimmunol.171.10.4969. [DOI] [PubMed] [Google Scholar]
  • [13].Combadiere B, Boissonnas A, Carcelain G, Lefranc E, Samri A, Bricaire F, et al. Distinct time effects of vaccination on long-term proliferative and IFN-gamma-producing T cell memory to smallpox in humans. J Exp Med. 2004 Jun 7;199(11):1585–93. doi: 10.1084/jem.20032083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Amanna IJ, Slifka MK, Crotty S. Immunity and immunological memory following smallpox vaccination. Immunol Rev. 2006 Jun;211:320–37. doi: 10.1111/j.0105-2896.2006.00392.x. [DOI] [PubMed] [Google Scholar]
  • [15].Frey SE, Newman FK, Yan L, Lottenbach KR, Belshe RB. Response to smallpox vaccine in persons immunized in the distant past. Jama. 2003 Jun 25;289(24):3295–9. doi: 10.1001/jama.289.24.3295. [DOI] [PubMed] [Google Scholar]
  • [16].Kim SH, Choi SJ, Park WB, Kim HB, Kim NJ, Oh MD, et al. Detailed kinetics of immune responses to a new cell culture-derived smallpox vaccine in vaccinia-naive adults. Vaccine. 2007 Aug 14;25(33):6287–91. doi: 10.1016/j.vaccine.2007.05.044. [DOI] [PubMed] [Google Scholar]
  • [17].Taub DD, Ershler WB, Janowski M, Artz A, Key ML, McKelvey J, et al. Immunity from smallpox vaccine persists for decades: a longitudinal study. The American journal of medicine. 2008 Dec;121(12):1058–64. doi: 10.1016/j.amjmed.2008.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Ferrier-Rembert A, Drillien R, Tournier JN, Garin D, Crance JM. Short- and long-term immunogenicity and protection induced by non-replicating smallpox vaccine candidates in mice and comparison with the traditional 1st generation vaccine. Vaccine. 2008 Mar 25;26(14):1794–804. doi: 10.1016/j.vaccine.2007.12.059. [DOI] [PubMed] [Google Scholar]
  • [19].Hopkins RJ, Lane JM. Clinical efficacy of intramuscular vaccinia immune globulin: a literature review. Clin Infect Dis. 2004 Sep 15;39(6):819–26. doi: 10.1086/422999. [DOI] [PubMed] [Google Scholar]
  • [20].Bray M, Martinez M, Smee DF, Kefauver D, Thompson E, Huggins JW. Cidofovir protects mice against lethal aerosol or intranasal cowpox virus challenge. J Infect Dis. 2000 Jan;181(1):10–9. doi: 10.1086/315190. [DOI] [PubMed] [Google Scholar]
  • [21].Buller RM, Owens G, Schriewer J, Melman L, Beadle JR, Hostetler KY. Efficacy of oral active ether lipid analogs of cidofovir in a lethal mousepox model. Virology. 2004 Jan 20;318(2):474–81. doi: 10.1016/j.virol.2003.11.015. [DOI] [PubMed] [Google Scholar]
  • [22].Smee DF, Bailey KW, Wong MH, Sidwell RW. Effects of cidofovir on the pathogenesis of a lethal vaccinia virus respiratory infection in mice. Antiviral Res. 2001 Oct;52(1):55–62. doi: 10.1016/s0166-3542(01)00159-0. [DOI] [PubMed] [Google Scholar]
  • [23].Whitley RJ. Smallpox: a potential agent of bioterrorism. Antiviral Res. 2003 Jan;57(12):7–12. doi: 10.1016/s0166-3542(02)00195-x. [DOI] [PubMed] [Google Scholar]
  • [24].Leitenberg M. Production and use of biological weapons: need for international sanctions? Jama. 1997 Aug 6;278(5):387–8. [PubMed] [Google Scholar]
  • [25].Guillemin J. Biological weapons and secrecy (WC 2300) Faseb J. 2005 Nov;19(13):1763–5. doi: 10.1096/fj.05-1102ufm. [DOI] [PubMed] [Google Scholar]
  • [26].Henderson DA. The looming threat of bioterrorism. Science. 1999 Feb 26;283(5406):1279–82. doi: 10.1126/science.283.5406.1279. [DOI] [PubMed] [Google Scholar]
  • [27].Fenner F. Smallpox and its eradication. World Health Organization; Geneva: 1988. [Google Scholar]
  • [28].Mack TM, Noble J, Jr., Thomas DB. A prospective study of serum antibody and protection against smallpox. Am J Trop Med Hyg. 1972 Mar;21(2):214–8. [Google Scholar]
  • [29].Sarkar JK, Mitra AC, Mukherjee MK. The minimum protective level of antibodies in smallpox. Bull World Health Organ. 1975;52(3):307–11. [PMC free article] [PubMed] [Google Scholar]
  • [30].Lane JM, Ruben FL, Abrutyn E, Millar JD. Deaths attributable to smallpox vaccination, 1959 to 1966, and 1968. Jama. 1970 Apr 20;212(3):441–4. [PubMed] [Google Scholar]
  • [31].Lane JM, Ruben FL, Neff JM, Millar JD. Complications of smallpox vaccination, 1968: results of ten statewide surveys. J Infect Dis. 1970 Oct;122(4):303–9. doi: 10.1093/infdis/122.4.303. [DOI] [PubMed] [Google Scholar]
  • [32].Halsell JS, Riddle JR, Atwood JE, Gardner P, Shope R, Poland GA, et al. Myopericarditis following smallpox vaccination among vaccinia-naive US military personnel. Jama. 2003 Jun 25;289(24):3283–9. doi: 10.1001/jama.289.24.3283. [DOI] [PubMed] [Google Scholar]
  • [33].Fulginiti VA, Papier A, Lane JM, Neff JM, Henderson DA. Smallpox vaccination: a review, part II. Adverse events. Clin Infect Dis. 2003 Jul 15;37(2):251–71. doi: 10.1086/375825. [DOI] [PubMed] [Google Scholar]
  • [34].Arita I. Smallpox vaccine and its stockpile in 2005. The Lancet infectious diseases. 2005 Oct;5(10):647–52. doi: 10.1016/S1473-3099(05)70242-5. [DOI] [PubMed] [Google Scholar]
  • [35].Smith BT, Inglesby TV, Brimmer E, Borio L, Franco C, Gronvall GK, et al. Navigating the storm: report and recommendations from the Atlantic Storm exercise. Biosecur Bioterror. 2005;3(3):256–67. doi: 10.1089/bsp.2005.3.256. [DOI] [PubMed] [Google Scholar]
  • [36].Neff J, Modlin J, Birkhead GS, Poland G, Robertson RM, Sepkowitz K, et al. Monitoring the safety of a smallpox vaccination program in the United States: report of the joint Smallpox Vaccine Safety Working Group of the advisory committee on immunization practices and the Armed Forces Epidemiological Board. Clin Infect Dis. 2008 Mar 15;46(Suppl 3):S258–70. doi: 10.1086/524749. [DOI] [PubMed] [Google Scholar]
  • [37].Poland GA, Grabenstein JD, Neff JM. The US smallpox vaccination program: a review of a large modern era smallpox vaccination implementation program. Vaccine. 2005 Mar 18;23(1718):2078–81. doi: 10.1016/j.vaccine.2005.01.012. [DOI] [PubMed] [Google Scholar]
  • [38].Rosenthal SR, Merchlinsky M, Kleppinger C, Goldenthal KL. Developing new smallpox vaccines. Emerging infectious diseases. 2001 Nov-Dec;7(6):920–6. doi: 10.3201/eid0706.010602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Frey SE, Couch RB, Tacket CO, Treanor JJ, Wolff M, Newman FK, et al. Clinical responses to undiluted and diluted smallpox vaccine. N Engl J Med. 2002 Apr 25;346(17):1265–74. doi: 10.1056/NEJMoa020534. [DOI] [PubMed] [Google Scholar]
  • [40].Talbot TR, Stapleton JT, Brady RC, Winokur PL, Bernstein DI, Germanson T, et al. Vaccination success rate and reaction profile with diluted and undiluted smallpox vaccine: a randomized controlled trial. Jama. 2004 Sep 8;292(10):1205–12. doi: 10.1001/jama.292.10.1205. [DOI] [PubMed] [Google Scholar]
  • [41].Gassmann R, Engler OB, Steffen R, Alex M, Czerny CP, Mutsch M. Clinical and immune response to undiluted and diluted smallpox vaccine. Swiss Med Wkly. 2008 Jul 12;138(2728):392–7. doi: 10.4414/smw.2008.11966. [DOI] [PubMed] [Google Scholar]
  • [42].Couch RB, Winokur P, Edwards KM, Black S, Atmar RL, Stapleton JT, et al. Reducing the dose of smallpox vaccine reduces vaccine-associated morbidity without reducing vaccination success rates or immune responses. J Infect Dis. 2007 Mar 15;195(6):826–32. doi: 10.1086/511828. [DOI] [PubMed] [Google Scholar]
  • [43].Hsieh SM, Chen SY, Sheu GC, Hung MN, Chou WH, Chang SC, et al. Clinical and immunological responses to undiluted and diluted smallpox vaccine with vaccinia virus of Lister strain. Vaccine. 2006 Jan 23;24(4):510–5. doi: 10.1016/j.vaccine.2005.07.082. [DOI] [PubMed] [Google Scholar]
  • [44].Gouvras G. Policies in place throughout the world: action by the European Union. Int J Infect Dis. 2004 Oct;8(Suppl 2):S21–30. doi: 10.1016/j.ijid.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • [45].Monath TP, Caldwell JR, Mundt W, Fusco J, Johnson CS, Buller M, et al. ACAM2000 clonal Vero cell culture vaccinia virus (New York City Board of Health strain)--a second-generation smallpox vaccine for biological defense. Int J Infect Dis. 2004 Oct;8(Suppl 2):S31–44. doi: 10.1016/j.ijid.2004.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Osborne JD, Da Silva M, Frace AM, Sammons SA, Olsen-Rasmussen M, Upton C, et al. Genomic differences of Vaccinia virus clones from Dryvax smallpox vaccine: the Dryvax-like ACAM2000 and the mouse neurovirulent Clone-3. Vaccine. 2007 Dec 17;25(52):8807–32. doi: 10.1016/j.vaccine.2007.10.040. [DOI] [PubMed] [Google Scholar]
  • [47].Frey SE, Newman FK, Kennedy JS, Ennis F, Abate G, Hoft DF, et al. Comparison of the safety and immunogenicity of ACAM1000, ACAM2000 and Dryvax((R)) in healthy vaccinia-naive adults. Vaccine. 2008 Dec 9; doi: 10.1016/j.vaccine.2008.11.079. [DOI] [PubMed] [Google Scholar]
  • [48].Artenstein AW, Johnson C, Marbury TC, Morrison D, Blum PS, Kemp T, et al. A novel, cell culture-derived smallpox vaccine in vaccinia-naive adults. Vaccine. 2005 May 9;23(25):3301–9. doi: 10.1016/j.vaccine.2005.01.079. [DOI] [PubMed] [Google Scholar]
  • [49].Marriott KA, Parkinson CV, Morefield SI, Davenport R, Nichols R, Monath TP. Clonal vaccinia virus grown in cell culture fully protects monkeys from lethal monkeypox challenge. Vaccine. 2008 Jan 24;26(4):581–8. doi: 10.1016/j.vaccine.2007.10.063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Greenberg RN, Kennedy JS. ACAM2000: a newly licensed cell culture-based live vaccinia smallpox vaccine. Expert Opin Investig Drugs. 2008 Apr;17(4):555–64. doi: 10.1517/13543784.17.4.555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Metzger W, Mordmueller BG. Vaccines for preventing smallpox. Cochrane database of systematic reviews (Online) 2007;(3) doi: 10.1002/14651858.CD004913.pub2. CD004913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Parrino J, Graham BS. Smallpox vaccines: Past, present, and future. J Allergy Clin Immunol. 2006 Dec;118(6):1320–6. doi: 10.1016/j.jaci.2006.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Poland GA. Smallpox vaccines: from first to second to third generation. Lancet. 2005 Jan 29 4;Feb 29 4;365(9457):362–3. doi: 10.1016/S0140-6736(05)17840-4. [DOI] [PubMed] [Google Scholar]
  • [54].Artenstein AW, Grabenstein JD. Smallpox vaccines for biodefense: need and feasibility. Expert Rev Vaccines. 2008 Oct;7(8):1225–37. doi: 10.1586/14760584.7.8.1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Murphy FA, Osburn BI. Adventitious agents and smallpox vaccine in strategic national stockpile. Emerging infectious diseases. 2005 Jul;11(7):1086–9. doi: 10.3201/eid1107.050277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Weltzin R, Liu J, Pugachev KV, Myers GA, Coughlin B, Blum PS, et al. Clonal vaccinia virus grown in cell culture as a new smallpox vaccine. Nat Med. 2003 Sep;9(9):1125–30. doi: 10.1038/nm916. [DOI] [PubMed] [Google Scholar]
  • [57].Greenberg RN, Kennedy JS, Clanton DJ, Plummer EA, Hague L, Cruz J, et al. Safety and immunogenicity of new cell-cultured smallpox vaccine compared with calf-lymph derived vaccine: a blind, single-centre, randomised controlled trial. Lancet. 2005 Jan 29 4;Feb 29 4;365(9457):398–409. doi: 10.1016/S0140-6736(05)17827-1. [DOI] [PubMed] [Google Scholar]
  • [58].Wiser I, Balicer RD, Cohen D. An update on smallpox vaccine candidates and their role in bioterrorism related vaccination strategies. Vaccine. 2007 Jan 22;25(6):976–84. doi: 10.1016/j.vaccine.2006.09.046. [DOI] [PubMed] [Google Scholar]
  • [59].Sugimoto M, Yamanouchi K. Characteristics of an attenuated vaccinia virus strain, LC16m0, and its recombinant virus vaccines. Vaccine. 1994 Jun;12(8):675–81. doi: 10.1016/0264-410x(94)90215-1. [DOI] [PubMed] [Google Scholar]
  • [60].Kidokoro M, Tashiro M, Shida H. Genetically stable and fully effective smallpox vaccine strain constructed from highly attenuated vaccinia LC16m8. Proc Natl Acad Sci U S A. 2005 Mar 15;102(11):4152–7. doi: 10.1073/pnas.0406671102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Empig C, Kenner JR, Perret-Gentil M, Youree BE, Bell E, Chen A, et al. Highly attenuated smallpox vaccine protects rabbits and mice against pathogenic orthopoxvirus challenge. Vaccine. 2006 Apr 24;24(17):3686–94. doi: 10.1016/j.vaccine.2005.03.029. [DOI] [PubMed] [Google Scholar]
  • [62].Mayr A, Stickl H, Muller HK, Danner K, Singer H. The smallpox vaccination strain MVA: marker, genetic structure, experience gained with the parenteral vaccination and behavior in organisms with a debilitated defence mechanism (author’s transl) Zentralbl Bakteriol [B] 1978 Dec;167(56):375–90. [PubMed] [Google Scholar]
  • [63].Mayr A. Historical review of smallpox, the eradication of smallpox and the attenuated smallpox MVA vaccine. Berl Munch Tierarztl Wochenschr. 1999 Sep;112(9):322–8. [PubMed] [Google Scholar]
  • [64].Vollmar J, Arndtz N, Eckl KM, Thomsen T, Petzold B, Mateo L, et al. Safety and immunogenicity of IMVAMUNE, a promising candidate as a third generation smallpox vaccine. Vaccine. 2006 Mar 15;24(12):2065–70. doi: 10.1016/j.vaccine.2005.11.022. [DOI] [PubMed] [Google Scholar]
  • [65].Kennedy JS, Greenberg RN. IMVAMUNE: modified vaccinia Ankara strain as an attenuated smallpox vaccine. Expert Rev Vaccines. 2009 Jan;8(1):13–24. doi: 10.1586/14760584.8.1.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Parrino J, McCurdy LH, Larkin BD, Gordon IJ, Rucker SE, Enama ME, et al. Safety, immunogenicity and efficacy of modified vaccinia Ankara (MVA) against Dryvax challenge in vaccinia-naive and vaccinia-immune individuals. Vaccine. 2007 Feb 9;25(8):1513–25. doi: 10.1016/j.vaccine.2006.10.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Tartaglia J, Perkus ME, Taylor J, Norton EK, Audonnet JC, Cox WI, et al. NYVAC: a highly attenuated strain of vaccinia virus. Virology. 1992 May;188(1):217–32. doi: 10.1016/0042-6822(92)90752-b. [DOI] [PubMed] [Google Scholar]
  • [68].Tartaglia J, Cox WI, Pincus S, Paoletti E. Safety and immunogenicity of recombinants based on the genetically-engineered vaccinia strain, NYVAC. Dev Biol Stand. 1994;82:125–9. [PubMed] [Google Scholar]
  • [69].Midgley CM, Putz MM, Weber JN, Smith GL. Vaccinia virus strain NYVAC induces substantially lower and qualitatively different human antibody responses compared with strains Lister and Dryvax. J Gen Virol. 2008 Dec;89(Pt 12):2992–7. doi: 10.1099/vir.0.2008/004440-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Paoletti E, Tartaglia J, Taylor J. Safe and effective poxvirus vectors--NYVAC and ALVAC. Dev Biol Stand. 1994;82:65–9. [PubMed] [Google Scholar]
  • [71].Edghill-Smith Y, Venzon D, Karpova T, McNally J, Nacsa J, Tsai WP, et al. Modeling a safer smallpox vaccination regimen, for human immunodeficiency virus type 1-infected patients, in immunocompromised macaques. J Infect Dis. 2003 Oct 15;188(8):1181–91. doi: 10.1086/378518. [DOI] [PubMed] [Google Scholar]
  • [72].Holzer GW, Remp G, Antoine G, Pfleiderer M, Enzersberger OM, Emsenhuber W, et al. Highly efficient induction of protective immunity by a vaccinia virus vector defective in late gene expression. J Virol. 1999 Jun;73(6):4536–42. doi: 10.1128/jvi.73.6.4536-4542.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Ober BT, Bruhl P, Schmidt M, Wieser V, Gritschenberger W, Coulibaly S, et al. Immunogenicity and safety of defective vaccinia virus lister: comparison with modified vaccinia virus Ankara. J Virol. 2002 Aug;76(15):7713–23. doi: 10.1128/JVI.76.15.7713-7723.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Coulibaly S, Bruhl P, Mayrhofer J, Schmid K, Gerencer M, Falkner FG. The nonreplicating smallpox candidate vaccines defective vaccinia Lister (dVV-L) and modified vaccinia Ankara (MVA) elicit robust long-term protection. Virology. 2005 Oct 10;341(1):91–101. doi: 10.1016/j.virol.2005.06.043. [DOI] [PubMed] [Google Scholar]
  • [75].Tagaya I, Amano H, Komatsu T, Uchida N, Kodama H. Supplement to the pathogenicity and immunogenicity of an attenuated vaccinia virus, strain DIs, in cynomolgus monkeys. Japanese journal of medical science & biology. 1974 Aug;27(4):215–28. doi: 10.7883/yoken1952.27.215. [DOI] [PubMed] [Google Scholar]
  • [76].Tagaya I, Amano H, Yuasa T. Improved plaque assay of a mutant of vaccinia virus, strain DIs, in chick embryo cell cultures. Japanese journal of medical science & biology. 1974 Aug;27(4):245–7. doi: 10.7883/yoken1952.27.245. [DOI] [PubMed] [Google Scholar]
  • [77].Ishii K, Ueda Y, Matsuo K, Matsuura Y, Kitamura T, Kato K, et al. Structural analysis of vaccinia virus DIs strain: application as a new replication-deficient viral vector. Virology. 2002 Oct 25;302(2):433–44. doi: 10.1006/viro.2002.1622. [DOI] [PubMed] [Google Scholar]
  • [78].Okamura T, Someya K, Matsuo K, Hasegawa A, Yamamoto N, Honda M. Recombinant vaccinia DIs expressing simian immunodeficiency virus gag and pol in mammalian cells induces efficient cellular immunity as a safe immunodeficiency virus vaccine candidate. Microbiology and immunology. 2006;50(12):989–1000. doi: 10.1111/j.1348-0421.2006.tb03867.x. [DOI] [PubMed] [Google Scholar]
  • [79].Ami Y, Izumi Y, Matsuo K, Someya K, Kanekiyo M, Horibata S, et al. Priming-boosting vaccination with recombinant Mycobacterium bovis bacillus Calmette-Guerin and a nonreplicating vaccinia virus recombinant leads to long-lasting and effective immunity. J Virol. 2005 Oct;79(20):12871–9. doi: 10.1128/JVI.79.20.12871-12879.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Mizutani T, Fukushi S, Ishii K, Sasaki Y, Kenri T, Saijo M, et al. Mechanisms of establishment of persistent SARS-CoV-infected cells. Biochem Biophys Res Commun. 2006 Aug 18;347(1):261–5. doi: 10.1016/j.bbrc.2006.06.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Buller RM, Smith GL, Cremer K, Notkins AL, Moss B. Decreased virulence of recombinant vaccinia virus expression vectors is associated with a thymidine kinase-negative phenotype. Nature. 1985 Oct 31 6;Nov 31 6;317(6040):813–5. doi: 10.1038/317813a0. [DOI] [PubMed] [Google Scholar]
  • [82].Vijaysri S, Jentarra G, Heck MC, Mercer AA, McInnes CJ, Jacobs BL. Vaccinia viruses with mutations in the E3L gene as potential replication-competent, attenuated vaccines: intra-nasal vaccination. Vaccine. 2008 Jan 30;26(5):664–76. doi: 10.1016/j.vaccine.2007.11.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Rodriguez D, Rodriguez JR, Rodriguez JF, Trauber D, Esteban M. Highly attenuated vaccinia virus mutants for the generation of safe recombinant viruses. Proc Natl Acad Sci U S A. 1989 Feb;86(4):1287–91. doi: 10.1073/pnas.86.4.1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Verardi PH, Jones LA, Aziz FH, Ahmad S, Yilma TD. Vaccinia virus vectors with an inactivated gamma interferon receptor homolog gene (B8R) are attenuated In vivo without a concomitant reduction in immunogenicity. J Virol. 2001 Jan;75(1):11–8. doi: 10.1128/JVI.75.1.11-18.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Zhu W, Fang Q, Zhuang K, Wang H, Yu W, Zhou J, et al. The attenuation of vaccinia Tian Tan strain by the removal of the viral M1L-K2L genes. Journal of virological methods. 2007 Sep;144(12):17–26. doi: 10.1016/j.jviromet.2007.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Legrand FA, Verardi PH, Jones LA, Chan KS, Peng Y, Yilma TD. Induction of potent humoral and cell-mediated immune responses by attenuated vaccinia virus vectors with deleted serpin genes. J Virol. 2004 Mar;78(6):2770–9. doi: 10.1128/JVI.78.6.2770-2779.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Dai K, Liu Y, Liu M, Xu J, Huang W, Huang X, et al. Pathogenicity and immunogenicity of recombinant Tiantan Vaccinia Virus with deleted C12L and A53R genes. Vaccine. 2008 Sep 15;26(39):5062–71. doi: 10.1016/j.vaccine.2008.06.011. [DOI] [PubMed] [Google Scholar]
  • [88].Reading PC, Smith GL. Vaccinia virus interleukin-18-binding protein promotes virulence by reducing gamma interferon production and natural killer and T-cell activity. J Virol. 2003 Sep;77(18):9960–8. doi: 10.1128/JVI.77.18.9960-9968.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Suter M, Meisinger-Henschel C, Tzatzaris M, Hulsemann V, Lukassen S, Wulff NH, et al. Modified vaccinia Ankara strains with identical coding sequences actually represent complex mixtures of viruses that determine the biological properties of each strain. Vaccine. 2009 Jun 17; doi: 10.1016/j.vaccine.2009.05.095. [DOI] [PubMed] [Google Scholar]
  • [90].Amies CR. Loss of immunogenic properties of vaccinia virus inactivated by formaldehyde. Can J Microbiol. 1961 Apr;7:141–52. doi: 10.1139/m61-019. [DOI] [PubMed] [Google Scholar]
  • [91].Turner GS, Squires EJ, Murray HG. Inactivated smallpox vaccine. A comparison of inactivation methods. J Hyg (Lond) 1970 Jun;68(2):197–210. doi: 10.1017/s0022172400028679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Madeley CR. The immunogenicity of heat-inactivated vaccinia virus in rabbits. J Hyg (Lond) 1968 Mar;66(1):89–107. doi: 10.1017/s0022172400040973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Boulter EA, Zwartouw HT, Titmuss DH, Maber HB. The nature of the immune state produced by inactivated vaccinia virus in rabbits. American journal of epidemiology. 1971 Dec;94(6):612–20. doi: 10.1093/oxfordjournals.aje.a121360. [DOI] [PubMed] [Google Scholar]
  • [94].Appleyard G, Hapel AJ, Boulter EA. An antigenic difference between intracellular and extracellular rabbitpox virus. J Gen Virol. 1971 Oct;13(1):9–17. doi: 10.1099/0022-1317-13-1-9. [DOI] [PubMed] [Google Scholar]
  • [95].Moss B. Poxvirus entry and membrane fusion. Virology. 2006 Jan 5;344(1):48–54. doi: 10.1016/j.virol.2005.09.037. [DOI] [PubMed] [Google Scholar]
  • [96].Smith GL, Vanderplasschen A, Law M. The formation and function of extracellular enveloped vaccinia virus. J Gen Virol. 2002 Dec;83(Pt 12):2915–31. doi: 10.1099/0022-1317-83-12-2915. [DOI] [PubMed] [Google Scholar]
  • [97].Galmiche MC, Goenaga J, Wittek R, Rindisbacher L. Neutralizing and protective antibodies directed against vaccinia virus envelope antigens. Virology. 1999 Feb 1;254(1):71–80. doi: 10.1006/viro.1998.9516. [DOI] [PubMed] [Google Scholar]
  • [98].Davies DH, McCausland MM, Valdez C, Huynh D, Hernandez JE, Mu Y, et al. Vaccinia virus H3L envelope protein is a major target of neutralizing antibodies in humans and elicits protection against lethal challenge in mice. J Virol. 2005 Sep;79(18):11724–33. doi: 10.1128/JVI.79.18.11724-11733.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Hooper JW, Custer DM, Schmaljohn CS, Schmaljohn AL. DNA vaccination with vaccinia virus L1R and A33R genes protects mice against a lethal poxvirus challenge. Virology. 2000 Jan 20;266(2):329–39. doi: 10.1006/viro.1999.0096. [DOI] [PubMed] [Google Scholar]
  • [100].Wolffe EJ, Vijaya S, Moss B. A myristylated membrane protein encoded by the vaccinia virus L1R open reading frame is the target of potent neutralizing monoclonal antibodies. Virology. 1995 Aug 1;211(1):53–63. doi: 10.1006/viro.1995.1378. [DOI] [PubMed] [Google Scholar]
  • [101].Hooper JW, Thompson E, Wilhelmsen C, Zimmerman M, Ichou MA, Steffen SE, et al. Smallpox DNA vaccine protects nonhuman primates against lethal monkeypox. J Virol. 2004 May;78(9):4433–43. doi: 10.1128/JVI.78.9.4433-4443.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Dean HJ, Haynes J, Schmaljohn C. The role of particle-mediated DNA vaccines in biodefense preparedness. Adv Drug Deliv Rev. 2005 Jun 17;57(9):1315–42. doi: 10.1016/j.addr.2005.01.012. [DOI] [PubMed] [Google Scholar]
  • [103].Fogg C, Lustig S, Whitbeck JC, Eisenberg RJ, Cohen GH, Moss B. Protective immunity to vaccinia virus induced by vaccination with multiple recombinant outer membrane proteins of intracellular and extracellular virions. J Virol. 2004 Oct;78(19):10230–7. doi: 10.1128/JVI.78.19.10230-10237.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [104].Xiao Y, Aldaz-Carroll L, Ortiz AM, Whitbeck JC, Alexander E, Lou H, et al. A protein-based smallpox vaccine protects mice from vaccinia and ectromelia virus challenges when given as a prime and single boost. Vaccine. 2007 Jan 26;25(7):1214–24. doi: 10.1016/j.vaccine.2006.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Hooper JW, Custer DM, Thompson E. Four-gene-combination DNA vaccine protects mice against a lethal vaccinia virus challenge and elicits appropriate antibody responses in nonhuman primates. Virology. 2003 Feb 1;306(1):181–95. doi: 10.1016/S0042-6822(02)00038-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Heraud JM, Edghill-Smith Y, Ayala V, Kalisz I, Parrino J, Kalyanaraman VS, et al. Subunit recombinant vaccine protects against monkeypox. J Immunol. 2006 Aug 15;177(4):2552–64. doi: 10.4049/jimmunol.177.4.2552. [DOI] [PubMed] [Google Scholar]
  • [107].Golden JW, Hooper JW. Heterogeneity in the A33 protein impacts the cross-protective efficacy of a candidate smallpox DNA vaccine. Virology. 2008 Jul 20;377(1):19–29. doi: 10.1016/j.virol.2008.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Aldaz-Carroll L, Xiao Y, Whitbeck JC, de Leon MP, Lou H, Kim M, et al. Major neutralizing sites on vaccinia virus glycoprotein B5 are exposed differently on variola virus ortholog B6. J Virol. 2007 Aug;81(15):8131–9. doi: 10.1128/JVI.00374-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Sakhatskyy P, Wang S, Zhang C, Chou TH, Kishko M, Lu S. Immunogenicity and protection efficacy of subunit-based smallpox vaccines using variola major antigens. Virology. 2008 Feb 5;371(1):98–107. doi: 10.1016/j.virol.2007.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Tscharke DC, Karupiah G, Zhou J, Palmore T, Irvine KR, Haeryfar SM, et al. Identification of poxvirus CD8+ T cell determinants to enable rational design and characterization of smallpox vaccines. J Exp Med. 2005 Jan 3;201(1):95–104. doi: 10.1084/jem.20041912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Mathew A, Terajima M, West K, Green S, Rothman AL, Ennis FA, et al. Identification of murine poxvirus-specific CD8+ CTL epitopes with distinct functional profiles. J Immunol. 2005 Feb 15;174(4):2212–9. doi: 10.4049/jimmunol.174.4.2212. [DOI] [PubMed] [Google Scholar]
  • [112].Jing L, Chong TM, Byrd B, McClurkan CL, Huang J, Story BT, et al. Dominance and diversity in the primary human CD4 T cell response to replication-competent vaccinia virus. J Immunol. 2007 May 15;178(10):6374–86. doi: 10.4049/jimmunol.178.10.6374. [DOI] [PubMed] [Google Scholar]
  • [113].Jing L, Chong TM, McClurkan CL, Huang J, Story BT, Koelle DM. Diversity in the acute CD8 T cell response to vaccinia virus in humans. J Immunol. 2005 Dec 1;175(11):7550–9. doi: 10.4049/jimmunol.175.11.7550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Snyder JT, Belyakov IM, Dzutsev A, Lemonnier F, Berzofsky JA. Protection against lethal vaccinia virus challenge in HLA-A2 transgenic mice by immunization with a single CD8+ T-cell peptide epitope of vaccinia and variola viruses. J Virol. 2004 Jul;78(13):7052–60. doi: 10.1128/JVI.78.13.7052-7060.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [115].Kennedy R, Poland GA. T-Cell epitope discovery for variola and vaccinia viruses. Rev Med Virol. 2007 Mar-Apr;17(2):93–113. doi: 10.1002/rmv.527. [DOI] [PubMed] [Google Scholar]
  • [116].Johnson KL, Ovsyannikova IG, Madden BJ, Poland GA, Muddiman DC. Accurate mass precursor ion data and tandem mass spectrometry identify a class I human leukocyte antigen A*0201-presented peptide originating from vaccinia virus. J Am Soc Mass Spectrom. 2005 Nov;16(11):1812–7. doi: 10.1016/j.jasms.2005.07.015. [DOI] [PubMed] [Google Scholar]
  • [117].Ovsyannikova IG, Johnson KL, Bergen HR, 3rd, Poland GA. Mass spectrometry and peptide-based vaccine development. Clinical pharmacology and therapeutics. 2007 Dec;82(6):644–52. doi: 10.1038/sj.clpt.6100389. [DOI] [PubMed] [Google Scholar]
  • [118].Moise L, McMurry JA, Buus S, Frey S, Martin WD, De Groot AS. In silico-accelerated identification of conserved and immunogenic variola/vaccinia T-cell epitopes. Vaccine. 2009 Jun 24; doi: 10.1016/j.vaccine.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [119].Clark TG, Cassidy-Hanley D. Recombinant subunit vaccines: potentials and constraints. Developments in biologicals. 2005;121:153–63. [PubMed] [Google Scholar]
  • [120].Bielinska AU, Chepurnov AA, Landers JJ, Janczak KW, Chepurnova TS, Luker GD, et al. A novel, killed-virus nasal vaccinia virus vaccine. Clin Vaccine Immunol. 2008 Feb;15(2):348–58. doi: 10.1128/CVI.00440-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [121].Golovkin M, Spitsin S, Andrianov V, Smirnov Y, Xiao Y, Pogrebnyak N, et al. Smallpox subunit vaccine produced in Planta confers protection in mice. Proc Natl Acad Sci U S A. 2007 Apr 17;104(16):6864–9. doi: 10.1073/pnas.0701451104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [122].Jahrling PB, Hensley LE, Martinez MJ, Leduc JW, Rubins KH, Relman DA, et al. Exploring the potential of variola virus infection of cynomolgus macaques as a model for human smallpox. Proc Natl Acad Sci U S A. 2004 Oct 19;101(42):15196–200. doi: 10.1073/pnas.0405954101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [123].Crowe JE., Jr. Genetic predisposition for adverse events after vaccination. J Infect Dis. 2007 Jul 15;196(2):176–7. doi: 10.1086/518800. [DOI] [PubMed] [Google Scholar]
  • [124].Stanley SL, Jr., Frey SE, Taillon-Miller P, Guo J, Miller RD, Koboldt DC, et al. The immunogenetics of smallpox vaccination. J Infect Dis. 2007 Jul 15;196(2):212–9. doi: 10.1086/518794. [DOI] [PubMed] [Google Scholar]
  • [125].Kennedy RB, Ovsyannikova IG, Pankratz VS, Vierkant RA, Jacobson RM, Ryan MA, et al. Gender effects on humoral immune responses to smallpox vaccine. Vaccine. 2009 May 26;27(2526):3319–23. doi: 10.1016/j.vaccine.2009.01.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Poland GA. Pharmacology, vaccinomics, and the second golden age of vaccinology. Clinical pharmacology and therapeutics. 2007 Dec;82(6):623–6. doi: 10.1038/sj.clpt.6100379. [DOI] [PubMed] [Google Scholar]
  • [127].Sakhatskyy P, Wang S, Chou TH, Lu S. Immunogenicity and protection efficacy of monovalent and polyvalent poxvirus vaccines that include the D8 antigen. Virology. 2006 Nov 25;355(2):164–74. doi: 10.1016/j.virol.2006.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Pulford DJ, Gates A, Bridge SH, Robinson JH, Ulaeto D. Differential efficacy of vaccinia virus envelope proteins administered by DNA immunisation in protection of BALB/c mice from a lethal intranasal poxvirus challenge. Vaccine. 2004 Sep 3;22(2526):3358–66. doi: 10.1016/j.vaccine.2004.02.034. [DOI] [PubMed] [Google Scholar]

RESOURCES