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 + 1992 word(s) 1992 2021-11-15 07:56:41 |
2 format correct Meta information modification 1992 2021-11-24 07:03:17 |

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.
Harun, N. Dengue Vaccine Development. Encyclopedia. Available online: https://encyclopedia.pub/entry/16327 (accessed on 26 April 2024).
Harun N. Dengue Vaccine Development. Encyclopedia. Available at: https://encyclopedia.pub/entry/16327. Accessed April 26, 2024.
Harun, Norshidah. "Dengue Vaccine Development" Encyclopedia, https://encyclopedia.pub/entry/16327 (accessed April 26, 2024).
Harun, N. (2021, November 24). Dengue Vaccine Development. In Encyclopedia. https://encyclopedia.pub/entry/16327
Harun, Norshidah. "Dengue Vaccine Development." Encyclopedia. Web. 24 November, 2021.
Dengue Vaccine Development
Edit

Approximately 100–400 million people from more than 100 countries in the tropical and subtropical world are affected by dengue infections. Recent scientific breakthroughs have brought new insights into novel strategies for the production of dengue antivirals and vaccines. The search for specific dengue inhibitors is expanding, and the mechanisms for evaluating the efficacy of novel drugs are currently established, allowing for expedited translation into human trials. Furthermore, in the aftermath of the only FDA-approved vaccine, Dengvaxia, a safer and more effective dengue vaccine candidate is making its way through the clinical trials. Until an effective licensed vaccine are available, disease monitoring and vector population control will be the mainstays of dengue prevention. In this review, we highlighted recent advances made in the perspectives of efforts made recently also to shed some light on the direction of the dengue vaccine development.

dengue virus NS2B/NS3pro drug discovery vaccine

1. Dengue Vaccine Development

In the development of dengue vaccines, a thorough understanding of immune responses to DENV aid in the formulation of an effective approach [1]. Live-attenuated vaccines, inactivated vaccines, recombinant subunit vaccines, and nucleic acid (DNA) vaccines are the primary forms of dengue vaccines currently under research (Figure 1).
Figure 1. Types of dengue vaccines.
These types of vaccines confer protection by increasing the immune responses to the E protein and non-structural protein 1 of the dengue virus (DENV) (NS1). The vaccine candidates that have progressed to the clinical trial stage are summarized in Table 1 below.
Table 1. DENV vaccines currently under development.
Vaccine Type Vaccine Name Developer Current Stage Target Antigen Strategy Key Clinical Outcome References
Live attenuated Dengvaxia© (CYD-TDV) Sanofi Pasteur Licensed Live virus DENV 1–4 genes substituted for the YF17D virus genes (prM/E). Age limit; increased risk of severe dengue in seronegative subjects but high effectiveness and safe in seropositive individuals [2][3][4]
Tetravax; TV003/TV005 NIH (USA); Butantan Institute (Brazil); Panacea Biotec Ltd. (India) Phase II/III Live virus Attenuation of DENV1, DENV3, DENV4, and a DENV2/DENV4 chimerical by excluding 30 nucleotides from the 3′ UTR. Well-tolerated; balanced immune response in subjects, effective with administration of a single dose. Adverse reaction (mild rash) [5][6]
TAK-003; DENVax Mahidol University; Inviragen; Takeda Phase III Live virus DENV2 PDK-53 attenuated vaccine coding sequences are replaced with DENV1, DENV3, and DENV4 coding sequences. Immunogenic and well-tolerated in multiple phase I and II clinical studies, independent of the participants’ age or serostatus, safety profile not entirely known [7]
TDEN F17/F19 WRAIR and GSK Phase II Live virus Involving primary cells of dog kidney (PDK) and lung cells of fetal rhesus (FrhL) in serial passages Proven to be a safe, well-tolerated, and immunogenic DENV vaccine candidate in phase II trial [8][9]
Inactivated TDEV-PIV GSK, Firocruz and WRAIR Merck Phase I Inactive virus Employing adjuvants and purified formalin-inactivated virus Well-tolerated, immunogenic in naive and seropositive individuals. No risk of re-activation and good immuno-logical balance [10][11]
Recombinant subunit V180 GSK, Firocruz and WRAIR Merck Phase I/II 80% of the E protein DEN-80E-containing recombinant truncated protein Induce steady immune responses against all DENV serotypes, decreasing the likelihood of the ADE effect [12][13]
Nucleic acid (DNA) TVDV U.S Naval Medical Research Centre Phase I prM and E proteins prM/E proteins are encoded via a recombinant plasmid vector Stable but lack of immunogenicity. Plasmid modification required. [14][15]
D1ME100 US Naval Medical Research Center Phase I prM and E proteins recombinant plasmid vector encoding prM/E No neutralizing antibody response detected in individuals with low-dose immunization [16]

2. Live-Attenuated Vaccines

Antigenic compounds synthesized from a living pathogen that has been engineered to be less virulent or avirulent are known as live-attenuated vaccines. Once administered, the viruses multiply locally, eliciting neutralizing antibodies and cell-mediated immune responses against the four dengue virus serotypes. These vaccines demonstrate the benefits of delivering protective antigens while offering long-term immune protection [17]. Using recombinant DNA technology, several live dengue attenuated vaccines have been developed.
Dengvaxia, also known as CYD-TDV, is the only approved tetravalent live-attenuated dengue vaccine candidate [2][3][4]. Receiving FDA approval in 2015, this vaccine is now accessible in more than 20 countries. Its usage has been allowed with strict restrictions on the recipients’ age and serostatus [18]. It offers high effectiveness in preventing dengue disease caused by DENV serotypes 1–4 and is safe in people who have had a past dengue infection, i.e., those who are seropositive. To date, the FDA has approved the usage in individuals 9 through 16 years of age with laboratory-confirmed previous dengue infection and living in endemic areas [19]. However, for those who are seronegative, the vaccine increases the chance of having severe dengue when the person has a spontaneous dengue illness about 3 years following immunization. Vaccination in the naive subjects stimulates the development of neutralizing antibodies against all four DENV serotypes. Specific antibodies against one or a few serotypes dominate this response, whereas reactions against the other serotypes are mostly due to cross-reactive antibodies. Moreover, it produces serotype-specific and cross-reactive T cell responses against DENV structural antigens. Therefore, the seronegative individuals may thus constitute a subclinical attenuated ‘primary-like’ illness. Vaccination in this group also results in different immunological effects depending on the serotypes. When compared to cross-protection evoked by vaccination in seropositive individuals, minimal cross-protection in seronegative individuals can be observed, posing a greater risk of inducing antibody-dependent enhancement (ADE) [20][21][22]. Consequently, WHO recommends that this vaccine is only given to seropositive individuals.
Tetravax (TV003/TV005), on the other hand, differs significantly from CYD-TDV in terms of the viral particle structure, infectivity, and immunogenicity [6][23]. To reduce DENV virulence, researchers utilized three untranslated regions (UTRs) deletions and structural gene prM/E chimerization. In comparison to TV003/TV005, CYD had a greater risk of viremia, lesser dengue virus type 2 resistance, and a reduced level of adaptive immune response [23].
TAK-003, often known as DENVax, is a live-attenuated chimeric tetravalent dengue vaccine [7]. At present, it is still in phase III of clinical investigations. The backbone of this vaccine is a weakened DENV2 strain (PDK-53) that contains the prM/E parts of all serotypes. The vaccine was proven to be immunogenic and well-tolerated in multiple phase I and II clinical studies, independent of the participants’ age or serostatus. TAK-003 showed a DENV serotype-dependent protective effectiveness, similar to its predecessor Dengvaxia, but with higher levels of DENV2 neutralizing antibodies and lower DENV3 and DENV4 protection rates, consequently, its safety profile is not entirely known [24]. Although a previous clinical study indicated it triggers CD8+ T lymphocytes directed at NS1, NS3, and NS5 in patients that have never been infected with DENV [25], this data were not included in later clinical studies.
In addition to live-attenuated dengue vaccines is TDEN F17/F19 [8][9]. In a phase II trial, this vaccine was proven to be a safe, well-tolerated, and immunogenic DENV vaccine candidate. One month following the second dosage, antibody responses to all four DENV types were recorded in more than half of the infants/toddlers and all of the children [26]. The vaccines used in these studies were lyophilized monovalent vaccines that were combined into a tetravalent vaccine at the point of administration.
Conclusively, based on the experiences obtained during the development prospective live-attenuated vaccines, the WHO have highlighted the guidelines on the quality, safety, and efficacy of the dengue tetravalent vaccines (live, attenuated). Furthermore, according to FDA, dengue live-attenuated vaccine, particularly Dengvaxia, elicits dengue-specific immune responses against the four dengue virus serotypes after injection. However, the exact mechanism of protection has yet to be discovered [19].

3. Inactivated Vaccines

Inactivated vaccines are antigenic compounds made up of denatured substances from other microbes such as bacteria and viruses that can provide protection against the live pathogen [27]. This vaccine stimulates immunity by using antigens from the capsid (c), membrane (M), envelope (E), and non-structural 1 (NS1) protein, although composite vaccinations provide superior protection compared to single-type immunizations.
The tetravalent purified formalin-inactivated virus (TPIV), which contains four non-active dengue serotypes, is an example of an inactivated vaccine now in clinical trials [28]. TDENV PIV/AS03B is now being investigated in a clinical study with various dosing regimens [10][11]. In both flavivirus-naive and experienced groups of infected individuals, TDEV PIV was well tolerated and immunogenic. Furthermore, this type of vaccine is safer than live-attenuated vaccines since there is no risk of reactivation and immunological balance is better regulated.

4. Recombinant Subunit Vaccine

Following the failure and controversy surrounding Dengvaxia®, recombinant subunit vaccination options have regained some interest. In this type of immunization, antigenic proteins produced by prokaryotic or eukaryotic cells generate long-lasting protective/therapeutic immune responses [29]. By far the most prevalent DENV recombinant subunit candidates are the envelope E protein or shortened variants. These rely on the formation of neutralizing antibodies to prevent DENV from infecting its host cells.
Despite the ease with which recombinant dengue proteins may be expressed in E. coli, there are potential concerns of being exposed to endotoxin contaminants and protein misfolding [30]. V180, which is made up of a shortened version of the protein DEN-80, is the most promising subunit vaccine [12][13]. Recombinant subunit vaccinations are more likely than live-attenuated vaccinations to induce steady immune responses against all DENV serotypes, decreasing the likelihood of the ADE effect [21].

5. DNA Vaccine

In the development of DNA vaccines, no less than a gene encoding specific antigens were incorporated into the plasmid. The E glycoprotein anchored to the prM protein mediates the first interaction between DENV and host cells, hence, it becomes the primary target for inducing neutralizing antibodies. Through in vivo injection of the expressed antigens, the vaccines induce both arms of the immune system which are the T cell responses and antibody production [31].
D1ME100 is an example of a DNA vaccine that has been clinically tested on Aotus nancymaae monkeys and humans [16][32]. In the initial phase of immunization, the vaccine proved to be safe and tolerable. Notwithstanding, with only about half of those who had high-dose immunization producing neutralizing antibodies, this immunogenicity produced was found to be weak. Furthermore, in those who received low-dose immunization, no neutralizing antibody response was detected [14]. Another example is the tetravalent dengue vaccine (TVDV), which is made up of four plasmids carrying the prM and E encoding genes from each DENV serotype [14][15]. The robust DENV-specific IFN T-cell response generated in 79% of the highest vaccination dosage recipients [15], however, is cause for concern. Despite its lack of immunogenicity, this vaccine has been shown to be stable, simple to make, inexpensive, and large-scale production. Plasmid modification with extremely efficient promoters, alternative delivery strategies, multiple doses, and co-immunization with adjuvants are recommended as solutions to the drawbacks of this form of vaccination.

6. An Ideal Vaccine against Dengue

Despite the fact that a dengue vaccine is now available, which is an important step forward, the long-lasting protective efficacy against each of the four dengue virus serotypes has yet to be confirmed. The characteristics of DENV, as well as the immunological protection and pathologic processes involved, including the transmission and epidemiology of dengue illness, have all hindered the development of a dengue vaccine. Vaccine development also has encountered conceptual problems since its inception, including the biology of the viruses that cause it, immunological protection and pathogenesis processes, and transmission and epidemiology.
Moreover, in endemic regions, the establishment of a protective antibody response against the infection is acquired through recurrent viral exposure. After being infected with one of the four DENV serotypes, the patient can be infected with any of the other serotypes. However, the infected individuals most likely do not manifest the clinical symptoms as a consequence of cross-protection.
The ideal dengue vaccine is abridged from the fact that it must elicit a multitypic response similar to that of people living in endemic areas, aided by persistent exposure and perhaps symptomatic or asymptomatic reinfection. An explicit elaboration on factors influencing vaccine responses, such as the pre-vaccination environment, as well as the significant challenges that face the development of an efficient/protective dengue vaccine, such as the presence of multiple serotypes, ADE, and cross-reactivity with other flaviviruses, have also been discussed to enhance the search of reliable dengue vaccine [33]. Hence, we concluded the key barriers to dengue vaccine development include a lack of a proper mechanistic investigation in pathogenesis and ADE.

References

  1. Wellekens, K.; Betrains, A.; De Munter, P.; Peetermans, W. Dengue: Current state one year before WHO 2010–2020 goals. Acta Clin. Belgica Int. J. Clin. Lab. Med. 2020, 1–9.
  2. Capeding, M.R.; Tran, N.H.; Hadinegoro, S.R.S.; Ismail, H.I.H.M.; Chotpitayasunondh, T.; Chua, M.N.; Luong, C.Q.; Rusmil, K.; Wirawan, D.N.; Nallusamy, R.; et al. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: A phase 3, randomised, observer-masked, placebo-controlled trial. Lancet 2014, 384, 1358–1365.
  3. Sabchareon, A.; Wallace, D.; Sirivichayakul, C.; Limkittikul, K.; Chanthavanich, P.; Suvannadabba, S.; Jiwariyavej, V.; Dulyachai, W.; Pengsaa, K.; Wartel, T.A.; et al. Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: A randomised, controlled phase 2b trial. Lancet 2012, 380, 1559–1567.
  4. Villar, L.; Dayan, G.H.; Arredondo-García, J.L.; Rivera, D.M.; Cunha, R.; Deseda, C.; Reynales, H.; Costa, M.S.; Morales-Ramírez, J.O.; Carrasquilla, G.; et al. Efficacy of a Tetravalent Dengue Vaccine in Children in Latin America. N. Engl. J. Med. 2015, 372, 113–123.
  5. Whitehead, S.S.; Durbin, A.P.; Pierce, K.K.; Elwood, D.; McElvany, B.D.; Fraser, E.A.; Carmolli, M.P.; Tibery, C.M.; Hynes, N.A.; Jo, M.; et al. In a randomized trial, the live attenuated tetravalent dengue vaccine TV003 is well-tolerated and highly immunogenic in subjects with flavivirus exposure prior to vaccination. PLoS Negl. Trop. Dis. 2017, 11, e0005584.
  6. Kirkpatrick, B.D.; Durbin, A.P.; Pierce, K.K.; Carmolli, M.P.; Tibery, C.M.; Grier, P.L.; Hynes, N.; Diehl, S.A.; Elwood, D.; Jarvis, A.P.; et al. Robust and Balanced Immune Responses to All 4 Dengue Virus Serotypes Following Administration of a Single Dose of a Live Attenuated Tetravalent Dengue Vaccine to Healthy, Flavivirus-Naive Adults. J. Infect. Dis. 2015, 212, 702–710.
  7. Sirivichayakul, C.; Barranco-Santana, E.A.; Esquilin-Rivera, I.; Oh, H.M.L.; Raanan, M.; Sariol, C.A.; Shek, L.P.; Simasathien, S.; Smith, M.K.; Velez, I.D.; et al. Safety and Immunogenicity of a Tetravalent Dengue Vaccine Candidate in Healthy Children and Adults in Dengue-Endemic Regions: A Randomized, Placebo-Controlled Phase 2 Study. J. Infect. Dis. 2016, 213, 1562–1572.
  8. Thomas, S.J.; Eckels, K.H.; Carletti, I.; De La Barrera, R.; Dessy, F.; Fernandez, S.; Putnak, R.; Toussaint, J.F.; Sun, W.; Bauer, K.; et al. A phase II, randomized, safety and immunogenicity study of a re-derived, live-attenuated dengue virus vaccine in healthy adults. Am. J. Trop. Med. Hyg. 2013, 88, 73–88.
  9. Bauer, K.; Esquilin, I.O.; Cornier, A.S.; Thomas, S.J.; Del Rio, A.I.Q.; Bertran-Pasarell, J.; Ramirez, J.O.M.; Diaz, C.; Carlo, S.; Eckels, K.H.; et al. A phase II, randomized, safety and immunogenicity trial of a re-derived, live-attenuated dengue virus vaccine in healthy children and adults living in puerto rico. Am. J. Trop. Med. Hyg. 2015, 93, 441–453.
  10. Schmidt, A.C.; Lin, L.; Martinez, L.J.; Ruck, R.C.; Eckels, K.H.; Collard, A.; De La Barrera, R.; Paolino, K.M.; Toussaint, J.F.; Lepine, E.; et al. Phase 1 randomized study of a tetravalent dengue purified inactivated vaccine in healthy adults in the United States. Am. J. Trop. Med. Hyg. 2017, 96, 1325–1337.
  11. Diaz, C.; Koren, M.; Lin, L.; Martinez, L.J.; Eckels, K.H.; Campos, M.; Jarman, R.G.; de la Barrera, R.; Lepine, E.; Febo, I.; et al. Safety and immunogenicity of different formulations of a tetravalent dengue purified inactivated vaccine in healthy adults from Puerto Rico: Final results after 3 years of follow-up from a randomized, placebo-controlled phase I study. Am. J. Trop. Med. Hyg. 2020, 102, 951–954.
  12. Coller, B.A.G.; Clements, D.E.; Bett, A.J.; Sagar, S.L.; Ter Meulen, J.H. The development of recombinant subunit envelope-based vaccines to protect against dengue virus induced disease. Vaccine 2011, 29, 7267–7275.
  13. Manoff, S.B.; Sausser, M.; Falk Russell, A.; Martin, J.; Radley, D.; Hyatt, D.; Roberts, C.C.; Lickliter, J.; Krishnarajah, J.; Bett, A.; et al. Immunogenicity and safety of an investigational tetravalent recombinant subunit vaccine for dengue: Results of a Phase I randomized clinical trial in flavivirus-naïve adults. Hum. Vaccines Immunother. 2019, 15, 2195–2204.
  14. Beckett, C.G.; Tjaden, J.; Burgess, T.; Danko, J.R.; Tamminga, C.; Simmons, M.; Wu, S.J.; Sun, P.; Kochel, T.; Raviprakash, K.; et al. Evaluation of a prototype dengue-1 DNA vaccine in a Phase 1 clinical trial. Vaccine 2011, 29, 960–968.
  15. Danko, J.R.; Kochel, T.; Teneza-Mora, N.; Luke, T.C.; Raviprakash, K.; Sun, P.; Simmons, M.; Moon, J.E.; De La Barrera, R.; Martinez, L.J.; et al. Safety and immunogenicity of a tetravalent dengue DNA vaccine administered with a cationic lipid-based adjuvant in a phase 1 clinical trial. Am. J. Trop. Med. Hyg. 2018, 98, 849–856.
  16. Kochel, T.J.; Raviprakash, K.; Hayes, C.G.; Watts, D.M.; Russell, K.L.; Gozalo, A.S.; Phillips, I.A.; Ewing, D.F.; Murphy, G.S.; Porter, K.R. A dengue virus serotype-1 DNA vaccine induces virus neutralizing antibodies and provides protection from viral challenge in Aotus monkeys. Vaccine 2000, 18, 3166–3173.
  17. Nivarthi, U.K.; Swanstrom, J.; Delacruz, M.J.; Patel, B.; Durbin, A.P.; Whitehead, S.S.; Kirkpatrick, B.D.; Pierce, K.K.; Diehl, S.A.; Katzelnick, L.; et al. A tetravalent live attenuated dengue virus vaccine stimulates balanced immunity to multiple serotypes in humans. Nat. Commun. 2021, 12, 1–12.
  18. Tully, D.; Griffiths, C.L. Dengvaxia: The world’s first vaccine for prevention of secondary dengue. Ther. Adv. Vaccines Immunother. 2021, 9, 251513552110158.
  19. FDA. Dengvaxia FDA 2019. Available online: https://www.fda.gov/vaccines-blood-biologics/dengvaxia (accessed on 26 October 2021).
  20. Guy, B.; Jackson, N. Dengue vaccine: Hypotheses to understand CYD-TDV-induced protection. Nat. Rev. Microbiol. 2015, 14, 45–54.
  21. Shukla, R.; Ramasamy, V.; Shanmugam, R.K.; Ahuja, R.; Khanna, N. Antibody-Dependent Enhancement: A Challenge for Developing a Safe Dengue Vaccine. Front. Cell. Infect. Microbiol. 2020, 10, 572681.
  22. Halstead, S.B.; Katzelnick, L.C.; Russell, P.K.; Markoff, L.; Aguiar, M.; Dans, L.R.; Dans, A.L. Ethics of a partially effective dengue vaccine: Lessons from the Philippines. Vaccine 2020, 38, 5572–5576.
  23. Whitehead, S.S. Development of TV003/TV005, a single dose, highly immunogenic live attenuated dengue vaccine; What makes this vaccine different from the Sanofi-Pasteur CYDTM vaccine? Expert Rev. Vaccines 2016, 15, 509–517.
  24. White, L.J.; Young, E.F.; Stoops, M.J.; Henein, S.R.; Adams, E.C.; Baric, R.S.; de Silva, A.M. Defining levels of dengue virus serotype-specific neutralizing antibodies induced by a live attenuated tetravalent dengue vaccine (Tak-003). PLoS Negl. Trop. Dis. 2021, 15, e0009258.
  25. Rivino, L.; Lim, M.Q. CD4+ and CD8+ T-cell immunity to Dengue—Lessons for the study of Zika virus. Immunology 2017, 150, 146–154.
  26. Watanaveeradej, V.; Gibbons, R.V.; Simasathien, S.; Nisalak, A.; Jarman, R.G.; Kerdpanich, A.; Tournay, E.; De La Barrerra, R.; Dessy, F.; Toussaint, J.F.; et al. Safety and immunogenicity of a rederived, live-attenuated dengue virus vaccine in healthy adults living in Thailand: A randomized trial. Am. J. Trop. Med. Hyg. 2014, 91, 119–128.
  27. Putnak, R.; Barvir, D.A.; Burrous, J.M.; Dubois, D.R.; D’Andrea, V.M.; Hoke, C.H.; Sadoff, J.C.; Eckels, K.H. Development of a purified, inactivated, dengue-2 virus vaccine prototype in Vero cells: Immunogenicity and protection in mice and rhesus monkeys. J. Infect. Dis. 1996, 174, 1176–1184.
  28. Prompetchara, E.; Ketloy, C.; Thomas, S.J.; Ruxrungtham, K. Dengue vaccine: Global development update. Asian Pac. J. Allergy Immunol. 2020, 38, 178–185.
  29. Tripathi, N.K.; Shrivastava, A. Recent Developments in Recombinant Protein–Based Dengue Vaccines. Front. Immunol. 2018, 9, 1919.
  30. Tripathi, N.K. Production and purification of recombinant proteins from Escherichia coli. ChemBioEng Rev. 2016, 3, 116–133.
  31. Azevedo, A.S.; Yamamura, A.M.Y.; Freire, M.S.; Trindade, G.F.; Bonaldo, M.; Galler, R.; Alves, A.M.B. DNA vaccines against dengue virus type 2 based on truncate envelope protein or its domain III. PLoS ONE 2011, 6, 20528.
  32. Maves, R.C.; Oré, R.M.C.; Porter, K.R.; Kochel, T.J. Immunogenicity and protective efficacy of a psoralen-inactivated dengue-1 virus vaccine candidate in Aotus nancymaae monkeys. Vaccine 2011, 29, 2691–2696.
  33. Izmirly, A.M.; Alturki, S.O.; Alturki, S.O.; Connors, J.; Haddad, E.K. Challenges in Dengue Vaccines Development: Pre-existing Infections and Cross-Reactivity. Front. Immunol. 2020, 11, 1055.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 305
Revisions: 2 times (View History)
Update Date: 29 Mar 2022
1000/1000