Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1878 2022-10-20 12:04:09 |
2 format correct + 20 word(s) 1898 2022-10-21 03:28:21 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Sah, R.;  Abdelaal, A.;  Reda, A.;  Mohanty, A.;  Lashin, B.I.;  Katamesh, B.E.;  Brakat, A.M.;  Al-Manaseer, B.M.;  Kaur, S.;  Asija, A.; et al. Vaccine and Monkeypox. Encyclopedia. Available online: https://encyclopedia.pub/entry/30507 (accessed on 20 May 2024).
Sah R,  Abdelaal A,  Reda A,  Mohanty A,  Lashin BI,  Katamesh BE, et al. Vaccine and Monkeypox. Encyclopedia. Available at: https://encyclopedia.pub/entry/30507. Accessed May 20, 2024.
Sah, Ranjit, Abdelaziz Abdelaal, Abdullah Reda, Aroop Mohanty, Basant Ismail Lashin, Basant E. Katamesh, Aml M. Brakat, Balqees Mahmoud Al-Manaseer, Sayanika Kaur, Ankush Asija, et al. "Vaccine and Monkeypox" Encyclopedia, https://encyclopedia.pub/entry/30507 (accessed May 20, 2024).
Sah, R.,  Abdelaal, A.,  Reda, A.,  Mohanty, A.,  Lashin, B.I.,  Katamesh, B.E.,  Brakat, A.M.,  Al-Manaseer, B.M.,  Kaur, S.,  Asija, A.,  Patel, N.K.,  Basnyat, S.,  Rabaan, A.A.,  Alhumaid, S.,  Albayat, H.,  Aljeldah, M.,  Shammari, B.R.A.,  Al-Najjar, A.H.,  Al-Jassem, A.K., ... Rodriguez-Morales, A.J. (2022, October 20). Vaccine and Monkeypox. In Encyclopedia. https://encyclopedia.pub/entry/30507
Sah, Ranjit, et al. "Vaccine and Monkeypox." Encyclopedia. Web. 20 October, 2022.
Vaccine and Monkeypox
Edit

The monkeypox virus (MPV) is a double-stranded DNA virus belonging to the Poxviridae family, Chordopoxvirinae subfamily, and Orthopoxvirus genus. It was called monkeypox because it was first discovered in monkeys, in a Danish laboratory, in 1958. Preventing the transmission and infection of MPV is associated with different challenges. The primary strategy of prevention would be vaccination. 

monkeypox vaccine outbreak

1. Monkeypox: The Past and Now

The monkeypox virus (MPV) is a double-stranded DNA virus belonging to the Poxviridae family, Chordopoxvirinae subfamily, and Orthopoxvirus genus [1]. It was called monkeypox because it was first discovered in monkeys, in a Danish laboratory, in 1958 [2]. However, the actual reservoir for MPV is still unknown [3]. On the other hand, the first reported case in humans was in an infant in the Democratic Republic of the Congo (DRC) in 1970 [4]. Then, it spread among other African countries, mainly Central and West Africa. Finally, in 2003, it started to spread outside Africa [5], reaching the United States by importing animals from Ghana [6]. Genetically, MPV has two clades: the West African and the Central African (Congo Basin) clade [7].
A systematic review showed that the number of monkeypox cases was highest in DRC, particularly in Central Africa, with 38, 520, and 85 confirmed cases reported during 1970–1979, 1990–1999, and 2010–2019, respectively. Outside Africa, mainly in the United States, MPV was detected in 47 cases from 2000–2009 [6]. That being said, MPV started re-emerging in 2022, affecting numerous patients with no connection worldwide. As of 3 July 2022, MPV was confirmed in 5783 cases in more than 52 nonendemic countries, with the highest numbers in the United Kingdom, Germany, Spain, France, the USA, and Portugal, in decreasing order [8].
This virus infects humans, causing monkeypox, which is quite similar to the disease caused by the smallpox virus. MPX has similar presentation and severity to smallpox but is associated with lower mortality (3–6% in the recent multicountry outbreak) and less human-to-human transmission [9][10]. Moreover, smallpox vaccines have been shown to provide cross-protection to MPV in 85% of cases [11]. As a result, the incidence rate of the MPV is highest among unvaccinated patients [6].
Current vaccines, although they provide cross-protection against monkeypox, are not specific for the causative virus and their efficacy in the light of the recent multicountry outbreak is still to be confirmed. In addition, as a consequence of the eradication and cessation of smallpox vaccination for four decades, MPV found an opportunity to re-emerge, but with different characteristics.

2. Prevention and Prophylaxis

Our immune system includes innate and adaptive components; innate immunity is the first defence line against infections, represented by physical, chemical, and microbiological barriers. Meanwhile, adaptive immunity comes after a previous infection to act specifically toward the previous immunogenic antigen [12]. In this regard, vaccines protect against such antigens without prior exposure, especially in prevalent infectious diseases.
One of the recent examples is the COVID-19 pandemic, which did not only affect the preparedness of the global healthcare system, but also affected it at the individual level, limiting the daily life activities of many people. The burden of COVID-19 pandemic was not reduced, in terms of associated morbidity and mortality, until effective vaccines were developed and disseminated. The herd immunity threshold level is calculated mathematically for each population; if this level is achieved, it will lead to herd immunity or population immunity, which protects unvaccinated individuals of the people indirectly when a certain percentage of the same population has been vaccinated and infected [13].
Herd immunity is a remarkable measure of vaccine efficacy. It helps to control and contain infectious diseases. However, the unknown mechanism of herd immunity led the researchers to understand its dynamics better to use it wisely in this era of newly emerging and re-emerging diseases [14], especially if the vaccines cannot cover the whole population for many reasons, including economic, religious, or medical reasons.

3. What Are the Different Types of Vaccines?

Live (-attenuated) vaccines are produced from weakened live viruses; they replicate in the host and create a milder form of the natural infection resulting in an immune response identical to natural infection. They do, however, have a slight risk of fatal infections when they replicate uncontrollably and, therefore, cannot be given to immunocompromised patients. There is also a risk of vertical and horizontal spread caused by contact with contaminated material [15]. The differences between live-attenuated and killed vaccines are highlighted in Table 1.
Table 1. The differences between live-attenuated and killed vaccines.
  Live Killed
Virus Weakened Live virus Entire virus is killed
Replication Can replicate and mimic natural infection Do not replicate
Immunity Greater and longer duration Lower and Shorter duration
Immune response Cell-mediated Humoral
Adjuvant Not needed Needed
Ig produced IgA and IgG IgG
Virulence May reverse No virulence
Booster Not required Required
Spread of strain Vertical and horizontal spread Not possible
Inactivated virus vaccines consist of whole viruses that have been treated with chemicals or heat. Chemicals, such as phenol and formaldehyde, and heat cause the denaturation of the surface proteins, inactivating them and making them noninfective. The treatment of the whole virus leaves some unchanged epitopes, which maintain some of their integrity and evoke an adaptive immune response by initiating antibody formation [16]. When injected, phagocytic immature dendritic cells divide the virus into smaller antigenic fragments, which are presented as antigenic fragments on the surface of MHC cells leading to the activation of B cells and T helper cells. Inactivated vaccines are safe with no adverse reactions. They are also noninfectious, heat-stable, and nontransmissible of the disease, as they cannot replicate. However, they are insufficient as a single dose and they usually do not provide immunity that is as strong as live vaccines; therefore, booster doses may be needed periodically to produce a sufficient immune response [17].
There are two types of mRNA vaccines: self-amplifying and nonreplicating. Self-amplifying vaccines contain codes for both the target antigen and the replicase complex, enabling the vaccine’s amplification. Nonreplicating RNA have codes for the target antigen alone and untranslated regions. Once inside the cell, the mRNA replicates to form a protein triggering an immune reaction and enabling the stimulation of both the innate and adaptive immune systems [18]. mRNA vaccines have many advantages; they can effectively activate both humoral and cell-mediated immunity, producing durable immune memory. Production of these vaccines is fast and inexpensive; thus, a large number can be made in a short time. In addition, they are stable, carry no risk of infection, do not integrate with the host genome, and are naturally degraded [19]. Noteworthily, a concern associated with this type of vaccine is the unknown risk for long-term adverse events [20].

4. Available Vaccines for Monkeypox Virus

Preventing the transmission and infection of MPV is associated with different challenges. The primary strategy of prevention would be vaccination. However, no specific vaccines have been developed for MPV. On the other hand, many previous studies conducted in the aftermath of smallpox infections demonstrated that smallpox vaccines could be effectively used to protect against the MPV [21]. For instance, older evidence indicates that smallpox vaccines might induce a cross-reaction with the MPV and can be efficacious as 85% in preventing the infection [22]. However, it should be noted that not all of these vaccines are primarily used because of their associated side effects and complications [23], which will be discussed in the following section.
Evidence indicates that remarkable advances have been introduced to vaccine technology after smallpox to enhance the safety and efficacy of these vaccines. Three generations of smallpox vaccines were developed accordingly with enhanced features [24]. For instance, first-generation smallpox vaccines were usually propagated through calfskin and collected at calf lymph. However, using these vaccines from the Smallpox Eradication Program (SEP) kept in national and WHO reserves is not currently recommended for MPV interventions. On the other hand, second-generation smallpox vaccines are produced with modern, sound manufacturing practices and propagated in tissue cell cultures. Accordingly, these vaccines have a reduced risk of contamination by surrounding factors.
Moreover, it has been shown that both of these generations are usually associated with an increased risk of developing adverse events as they have a replication-competent vaccinia virus [25][26]. Third-generation smallpox vaccines are manufactured similarly to second-generation ones. However, they have enhanced safety profiles because they contain attenuated vaccinia viruses rather than replication-competent vaccinia ones [5][27][28]Table 2 summarises the structure and efficacy of currently available vaccines to help prevent MPV infection.
Table 2. Structures, generations, and effectiveness of reported smallpox vaccines for preventing monkeypox infections.
Vaccine Generation Effective for Use for
Monkeypox
Structure Injection
Materials
Presentations
LC16 3rd generation Infants, children, and adults (all ages) No Minimally replicating vaccinia virus Bifurcated needle Freeze-dried Multidose vials
ACAM2000 2nd generation Adults (18–64) USA: postexposure prophylaxis Propagated in tissue cell culture and produced under good manufacturing practices (live, replication-competent virus) Bifurcated needle Freeze-dried Multidose vials
JYNNEOSTM/MVA-BN 3rd generation General adult population Yes (USA, UK, Canada) Nonreplicating vaccinia virus Needle and syringe (subcutaneous administration) Liquid frozen or lyophilised (freeze-dried) Single-dose vials (Multidose vials possible)
Vaccinia (Dryvax, Lister, Copenhagen) 1st generation - No Several different strains of vaccinia virus propagated in calf lymph (live, replication-competent virus) Bifurcated needle Liquid frozen or lyophilised vials or ampoules
The Food and Drug Administration approved the second-generation smallpox vaccine ACAM2000 to be used in emergencies and outbreaks of smallpox for postexposure prophylaxis. Accordingly, it has been purchased for the Strategic National Stockpile (SNS) and was used for different population groups [1]. Furthermore, in 2019, MVA-BN or JYNNEOSTM was approved in the United States and Canada after various animal studies, and clinical trials showed the high efficacy and safety of this vaccine, which can also be used in different population groups for the prevention of MPV infection [2][29][30][31][32][33][34][35][36]. Furthermore, JYNNEOSTM was approved based on the data suggesting its noninferior immunogenicity to other smallpox vaccines, its efficacy in animals, and its safety profile in humans [27][37][38][39][40][41]. Moreover, LC16 has been approved in the United States by the emergency investigational new drug program of the US Food and Drug Administration and in Japan, secondary to the reported protective effects in animal models and immunogenicity in human studies [41][42][43]. Although LC16 is the only approved smallpox vaccine for use in children, there are no data on its efficacy in preventing MPV infection.
It should be noted that these vaccines are usually efficacious in preventing MPV infection when used as pre-exposure approaches. Nevertheless, experts demonstrated that postexposure vaccination could also intervene against the development of severe diseases or reduce the severity of the conditions of infected MPV patients [44]. In this context, it can be suggested that it is better to be vaccinated sooner after exposure. Evidence from the Centers for Disease Control and Prevention (CDC) recommends that vaccinating exposed individuals should be conducted within four days of exposure to prevent the onset of the disease [45]. Therefore, the beginning of the illness may be inevitable in individuals that did not receive the vaccination within this period. However, vaccination within the first two weeks might intervene against developing the severe disease [46].
There is also evidence that the protective efficacy of smallpox vaccines usually fades over time. However, it has been shown that postvaccination protection might last for up to 20 years. Besides, although the protective efficacy of the smallpox vaccine fades with time, vaccinated individuals will still have lifelong protection against developing a severe disease secondary to the presence of memory B and T cells [41][47]. Therefore, immunity against the MPV should be expected in previously vaccinated individuals against smallpox [48]. The CDC also recommends that vaccination be given to individuals exposed to the MPV and not vaccinated within the last three years [45]. In this context, it has been demonstrated that receiving the smallpox vaccine as soon as possible would be more effective in preventing MPV infection [49].

References

  1. Petersen, B.W.; Harms, T.J.; Reynolds, M.G.; Harrison, L.H. Use of Vaccinia Virus Smallpox Vaccine in Laboratory and Health Care Personnel at Risk for Occupational Exposure to Orthopoxviruses—Recommendations of the Advisory Committee on Immunization Practices (ACIP), 2015. Morb. Mortal. Wkly. Rep. 2016, 65, 257–262.
  2. von Krempelhuber, A.; Vollmar, J.; Pokorny, R.; Rapp, P.; Wulff, N.; Petzold, B.; Handley, A.; Mateo, L.; Siersbol, H.; Kollaritsch, H.; et al. A randomized, double-blind, dose-finding Phase II study to evaluate immunogenicity and safety of the third generation smallpox vaccine candidate IMVAMUNE. Vaccine 2010, 28, 1209–1216.
  3. Reynolds, M.G.; Guagliardo, S.A.J.; Nakazawa, Y.J.; Doty, J.B.; Mauldin, M.R. Understanding orthopoxvirus host range and evolution: From the enigmatic to the usual suspects. Curr. Opin. Virol. 2018, 28, 108–115.
  4. Breman, J.G.; Steniowski, M.; Zanotto, E.; Gromyko, A.; Arita, I. Human monkeypox, 1970–1979. Bull. World Health Organ. 1980, 58, 165–182.
  5. Zitzmann-Roth, E.M.; von Sonnenburg, F.; de la Motte, S.; Arndtz-Wiedemann, N.; von Krempelhuber, A.; Uebler, N.; Vollmar, J.; Virgin, G.; Chaplin, P. Cardiac safety of Modified Vaccinia Ankara for vaccination against smallpox in a young, healthy study population. PLoS ONE 2015, 10, e0122653.
  6. Bunge, E.M.; Hoet, B.; Chen, L.; Lienert, F.; Weidenthaler, H.; Baer, L.R.; Steffen, R. The changing epidemiology of human monkeypox—A potential threat? A systematic review. PLoS Negl. Trop. Dis. 2022, 16, e0010141.
  7. Likos, A.M.; Sammons, S.A.; Olson, V.A.; Frace, A.M.; Li, Y.; Olsen-Rasmussen, M.; Davidson, W.; Galloway, R.; Khristova, M.L.; Reynolds, M.G. A tale of two clades: Monkeypox viruses. J. Gen. Virol. 2005, 86, 2661–2672.
  8. Monkeypox Cases Worldwide: Key Statistics. Available online: http://www.global.health (accessed on 26 June 2022).
  9. Shafaati, M.; Zandi, M. Monkeypox virus neurological manifestations in comparison to other orthopoxviruses. Travel Med. Infect. Dis. 2022, 49, 102414.
  10. Shapovalova, V. Monkeypox Virus–New Challenges of Modernity: Experimental Organizational and Legal, Clinical and Pharmacological Studies. SSP Mod. Pharm. Med. 2022, 2, 1–15.
  11. Fine, P.; Jezek, Z.; Grab, B.; Dixon, H. The transmission potential of monkeypox virus in human populations. Int. J. Epidemiol. 1988, 17, 643–650.
  12. Jacqueline, P.; Bryony, C. An overview of the immune system. Lancet 2001, 357, 1777–1789.
  13. Chaudhary, J.K.; Yadav, R.; Chaudhary, P.K.; Maurya, A.; Kant, N.; Rugaie, O.A.; Haokip, H.R.; Yadav, D.; Roshan, R.; Prasad, R.; et al. Insights into COVID-19 Vaccine Development Based on Immunogenic Structural Proteins of SARS-CoV-2, Host Immune Responses Herd Immunity. Cells 2021, 10, 2949.
  14. Peters, M.A. The coming pandemic era. Educ. Philos. Theory 2022, 54, 656–661.
  15. Cohen, N.D.; Bordin, A.I. Principles of Vaccination. In Equine Clinical Immunology; John Wiley & Sons, Inc.: Chichester, UK, 2015; pp. 263–278.
  16. Siegriest, C.A. Vaccine Immunology. In Vaccines; Elsevier: Amsterdam, The Netherlands, 2008.
  17. Baxter, D. Active and passive immunity, vaccine types, excipients and licensing. Occup. Med. 2007, 57, 552–556.
  18. Maruggi, G.; Zhang, C.; Li, J.; Ulmer, J.B.; Yu, D. mRNA as a transformative technology for vaccine development to control infectious diseases. Mol. Ther. 2019, 27, 757–772.
  19. Park, J.W.; Lagniton, P.N.; Liu, Y.; Xu, R.-H. mRNA vaccines for COVID-19: What, why and how. Int. J. Biol. Sci. 2021, 17, 1440–1466.
  20. Hitti, F.L.; Weissman, D. Debunking mRNA Vaccine Misconceptions-An Overview for Medical Professionals. Am. J. Med. 2021, 134, 703–704.
  21. Jezek, Z.; Grab, B.; Szczeniowski, M.V.; Paluku, K.M.; Mutombo, M. Human monkeypox: Secondary attack rates. Bull. World Health Organ. 1988, 66, 465–470.
  22. McCollum, A.M.; Damon, I.K. Human monkeypox. Clinical infectious diseases: An official publication of the Infectious Diseases Society of America. Clin. Infect. Dis. 2014, 58, 260–267.
  23. Rimoin, A.W.; Graham, B.S. Whither monkeypox vaccination. Vaccine 2011, 29 (Suppl. 4), D60–D64.
  24. World Health Organization. Vaccines and Immunization for Monkeypox: Interim Guidance; World Health Organization: Geneva, Switzerland, 2022.
  25. Berhanu, A.; Prigge, J.T.; Silvera, P.M.; Honeychurch, K.M.; Hruby, D.E.; Grosenbach, D.W. Treatment with the smallpox antiviral tecovirimat (ST-246) alone or in combination with ACAM2000 vaccination is effective as a postsymptomatic therapy for monkeypox virus infection. Antimicrob. Agents Chemother. 2015, 59, 4296–4300.
  26. Faix, D.J.; Gordon, D.M.; Perry, L.N.; Raymond-Loher, I.; Tati, N.; Lin, G.; DiPietro, G.; Selmani, A.; Decker, M.D. Prospective safety surveillance study of ACAM2000 smallpox vaccine in deploying military personnel. Vaccine 2020, 38, 7323–7330.
  27. Jackson, L.A.; Frey, S.E.; El Sahly, H.M.; Mulligan, M.J.; Winokur, P.L.; Kotloff, K.L.; Campbell, J.D.; Atmar, R.L.; Graham, I.; Anderson, E.J.; et al. Safety and immunogenicity of a modified vaccinia Ankara vaccine using three immunization schedules and two modes of delivery: A randomized clinical non-inferiority trial. Vaccine 2017, 35, 1675–1682.
  28. Greenberg, R.N.; Hurley, M.Y.; Dinh, D.V.; Mraz, S.; Vera, J.G.; von Bredow, D.; von Krempelhuber, A.; Roesch, S.; Virgin, G.; Arndtz-Wiedemann, N.; et al. A Multicenter, Open-Label, Controlled Phase II Study to Evaluate Safety and Immunogenicity of MVA Smallpox Vaccine (IMVAMUNE) in 18–40 Year Old Subjects with Diagnosed Atopic Dermatitis. PLoS ONE 2015, 10, e0138348.
  29. Stittelaar, K.J.; van Amerongen, G.; Kondova, I.; Kuiken, T.; van Lavieren, R.F.; Pistoor, F.H.; Niesters, H.G.; van Doornum, G.; van der Zeijst, B.A.; Mateo, L.; et al. Modified vaccinia virus Ankara protects macaques against respiratory challenge with monkeypox virus. J. Virol. 2005, 79, 7845–7851.
  30. Keckler, M.S.; Carroll, D.S.; Gallardo-Romero, N.F.; Lash, R.R.; Salzer, J.S.; Weiss, S.L.; Patel, N.; Clemmons, C.J.; Smith, S.K.; Hutson, C.L.; et al. Establishment of the black-tailed prairie dog (Cynomys ludovicianus) as a novel animal model for comparing smallpox vaccines administered preexposure in both high- and low-dose monkeypox virus challenges. J. Virol. 2011, 85, 7683–7698.
  31. Earl, P.L.; Americo, J.L.; Wyatt, L.S.; Espenshade, O.; Bassler, J.; Gong, K.; Lin, S.; Peters, E.; Rhodes, L., Jr.; Spano, Y.E.; et al. Rapid protection in a monkeypox model by a single injection of a replication-deficient vaccinia virus. Proc. Natl. Acad. Sci. USA 2008, 105, 10889–10894.
  32. Walsh, S.R.; Wilck, M.B.; Dominguez, D.J.; Zablowsky, E.; Bajimaya, S.; Gagne, L.S.; Verrill, K.A.; Kleinjan, J.A.; Patel, A.; Zhang, Y.; et al. Safety and immunogenicity of modified vaccinia Ankara in hematopoietic stem cell transplant recipients: A randomized, controlled trial. J. Infect. Dis. 2013, 207, 1888–1897.
  33. Vollmar, J.; Arndtz, N.; Eckl, K.M.; Thomsen, T.; Petzold, B.; Mateo, L.; Schlereth, B.; Handley, A.; King, L.; Hülsemann, V.; et al. Safety and immunogenicity of IMVAMUNE, a promising candidate as a third generation smallpox vaccine. Vaccine 2006, 24, 2065–2070.
  34. Greenberg, R.N.; Overton, E.T.; Haas, D.W.; Frank, I.; Goldman, M.; von Krempelhuber, A.; Virgin, G.; Bädeker, N.; Vollmar, J.; Chaplin, P. Safety, immunogenicity, and surrogate markers of clinical efficacy for modified vaccinia Ankara as a smallpox vaccine in HIV-infected subjects. J. Infect. Dis. 2013, 207, 749–758.
  35. Frey, S.E.; Winokur, P.L.; Salata, R.A.; El-Kamary, S.S.; Turley, C.B.; Walter, E.B., Jr.; Hay, C.M.; Newman, F.K.; Hill, H.R.; Zhang, Y.; et al. Safety and immunogenicity of IMVAMUNE® smallpox vaccine using different strategies for a post event scenario. Vaccine 2013, 31, 3025–3033.
  36. Frey, S.E.; Newman, F.K.; Kennedy, J.S.; Sobek, V.; Ennis, F.A.; Hill, H.; Yan, L.K.; Chaplin, P.; Vollmar, J.; Chaitman, B.R.; et al. Clinical and immunologic responses to multiple doses of IMVAMUNE (Modified Vaccinia Ankara) followed by Dryvax challenge. Vaccine 2007, 25, 8562–8573.
  37. Overton, E.T.; Lawrence, S.J.; Stapleton, J.T.; Weidenthaler, H.; Schmidt, D.; Koenen, B.; Silbernagl, G.; Nopora, K.; Chaplin, P. A randomized phase II trial to compare safety and immunogenicity of the MVA-BN smallpox vaccine at various doses in adults with a history of AIDS. Vaccine 2020, 38, 2600–2607.
  38. Pittman, P.R.; Hahn, M.; Lee, H.S.; Koca, C.; Samy, N.; Schmidt, D.; Hornung, J.; Weidenthaler, H.; Heery, C.R.; Meyer, T.P.H.; et al. Phase 3 Efficacy Trial of Modified Vaccinia Ankara as a Vaccine against Smallpox. N. Engl. J. Med. 2019, 381, 1897–1908.
  39. Overton, E.T.; Lawrence, S.J.; Wagner, E.; Nopora, K.; Rösch, S.; Young, P.; Schmidt, D.; Kreusel, C.; De Carli, S.; Meyer, T.P.; et al. Immunogenicity and safety of three consecutive production lots of the non replicating smallpox vaccine MVA: A randomised, double blind, placebo controlled phase III trial. PLoS ONE 2018, 13, e0195897.
  40. Greenberg, R.N.; Hay, C.M.; Stapleton, J.T.; Marbury, T.C.; Wagner, E.; Kreitmeir, E.; Röesch, S.; von Krempelhuber, A.; Young, P.; Nichols, R.; et al. A Randomized, Double-Blind, Placebo-Controlled Phase II Trial Investigating the Safety and Immunogenicity of Modified Vaccinia Ankara Smallpox Vaccine (MVA-BN®) in 56–80-Year-Old Subjects. PLoS ONE 2016, 11, e0157335.
  41. Overton, E.T.; Stapleton, J.; Frank, I.; Hassler, S.; Goepfert, P.A.; Barker, D.; Wagner, E.; von Krempelhuber, A.; Virgin, G.; Meyer, T.P.; et al. Safety and Immunogenicity of Modified Vaccinia Ankara-Bavarian Nordic Smallpox Vaccine in Vaccinia-Naive and Experienced Human Immunodeficiency Virus-Infected Individuals: An Open-Label, Controlled Clinical Phase II Trial. Open Forum Infect. Dis. 2015, 2, ofv040.
  42. Nishiyama, Y.; Fujii, T.; Kanatani, Y.; Shinmura, Y.; Yokote, H.; Hashizume, S. Freeze-dried live attenuated smallpox vaccine prepared in cell culture “LC16-KAKETSUKEN”: Post-marketing surveillance study on safety and efficacy compliant with Good Clinical Practice. Vaccine 2015, 33, 6120–6127.
  43. Kennedy, J.S.; Gurwith, M.; Dekker, C.L.; Frey, S.E.; Edwards, K.M.; Kenner, J.; Lock, M.; Empig, C.; Morikawa, S.; Saijo, M.; et al. Safety and immunogenicity of LC16m8, an attenuated smallpox vaccine in vaccinia-naive adults. J. Infect. Dis. 2011, 204, 1395–1402.
  44. (CDC) Center for Disease Control and Prevention. Monkeypox—Treatment. Available online: https://www.cdc.gov/poxvirus/monkeypox/treatment.html (accessed on 13 June 2022).
  45. (CDC) Center for Disease Control and Prevention. Monkeypox and Smallpox Vaccine Guidance. Available online: https://www.cdc.gov/poxvirus/monkeypox/clinicians/smallpox-vaccine.html (accessed on 13 June 2022).
  46. Petersen, B.W.; Kabamba, J.; McCollum, A.M.; Lushima, R.S.; Wemakoy, E.O.; Muyembe Tamfum, J.J.; Nguete, B.; Hughes, C.M.; Monroe, B.P.; Reynolds, M.G. Vaccinating against monkeypox in the Democratic Republic of the Congo. Antivir. Res. 2019, 162, 171–177.
  47. Darsow, U.; Sbornik, M.; Rombold, S.; Katzer, K.; von Sonnenburg, F.; Behrendt, H.; Ring, J. Long-term safety of replication-defective smallpox vaccine (MVA-BN) in atopic eczema and allergic rhinitis. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 1971–1977.
  48. Kunasekaran, M.P.; Chen, X.; Costantino, V.; Chughtai, A.A.; MacIntyre, C.R. Evidence for Residual Immunity to Smallpox After Vaccination and Implications for Re-emergence. Mil. Med. 2019, 184, e668–e679.
  49. Frey, S.E.; Wald, A.; Edupuganti, S.; Jackson, L.A.; Stapleton, J.T.; El Sahly, H.; El-Kamary, S.S.; Edwards, K.; Keyserling, H.; Winokur, P.; et al. Comparison of lyophilized versus liquid modified vaccinia Ankara (MVA) formulations and subcutaneous versus intradermal routes of administration in healthy vaccinia-naïve subjects. Vaccine 2015, 33, 5225–5234.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , , , , , , , , , , , , , , , , ,
View Times: 520
Revisions: 2 times (View History)
Update Date: 24 Oct 2022
1000/1000
Video Production Service