Articles Service
Minireview
Current Status of Epidemiology, Diagnosis, Therapeutics, and Vaccines for the Re-Emerging Human Monkeypox Virus
Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
Correspondence to:J. Microbiol. Biotechnol. 2023; 33(8): 981-991
Published August 28, 2023 https://doi.org/10.4014/jmb.2306.06033
Copyright © The Korean Society for Microbiology and Biotechnology.
Abstract
Keywords
Graphical Abstract
Introduction
Monkeypox (Mpox), a virus-borne infectious disease that recently re-emerged worldwide, has raised concerns. It exhibits symptoms similar to those of smallpox, such as high fever, generalized body aches, and the development of distinctive rashes and pustules on the skin that last a few weeks [1]. It is caused by infection with Mpox virus (MPXV), which belongs to the
MPXV is a large virus (200–250 nm in diameter) with a lipid envelope and a linear double-stranded DNA genome of approximately 197 kbp. The viral genome contains about 6.4 kbp inverted terminal repeats (ITR) and covalently closed hairpin ends. Genetic diversity at both ends affects the virulence, pathogenicity, and fatality of
The MPXV genome contains approximately 190 ORFs. However, the roles of many proteins, excluding certain key viral proteins involved in viral replication and assembly, are not yet clearly understood, although they are thought to be involved in host tropism and immunomodulation. The genome has a low GC content of approximately 33% and is characterized by the presence of numerous local tandem repeats, including the ITR. These features make it challenging to achieve a consistent reconstruction during gene replication and virion assembly [5].
MPXV is composed of several constituents (genomic DNA, nucleocapsid, core, lateral body, inner membrane, envelope, and surface proteins) (Fig. 1A). The core is dumbbell-shaped and is surrounded by the core membrane, palisade layer, and lateral bodies.
-
Fig. 1. An overview of MPXV structure and lifecycle.
(A) The virion mainly consists of DNA genome, nucleocapsids, dumbbell-shaped core, lateral bodies, inner membrane, envelope, and surface proteins. There are at least 25 species of surface proteins, which are distributed on envelope and/or inner membrane, respectively. (B) The attachment and membrane fusion of virions are dependent on the interaction among viral surface proteins, extracellular GAGs, and matrix proteins of the host cell. Uncoated genomic DNA immediately forms concentrated complex known as DNA factory to undergo the stages of transcription, replication, and translation, leading to the assembly of IV. Subsequently, morphogenesis occurs in the cytoplasm to shape IV into IMV. EVs with envelope formed in the Golgi complex are released from the cells through exocytosis, and IMVs without envelope can also be released from the cells through cell lysis. GAG, glycosaminoglycan; IV, immature virion; IMV, intracellular mature virion; IEV, intracellular enveloped virion; EEV, extracellular enveloped virion.
MPXV entry occurs in a manner similar to the distinctive entry mechanism of other poxviruses; however, the detailed mechanism has not yet been fully elucidated. Herein, we briefly describe the basic entry process of vaccinia virus (VACV), which is a well-known poxvirus relatively, and the viral surface proteins involved, as well as the host receptors. It has been known that poxvirus has over 8 surface proteins on the envelope and over 20 surface proteins on the inner membrane. Adhesion to cellular receptors, membrane fusion, and inter-host transmission occur through parallel interactions between these surface proteins and their respective host receptors. Fortunately, surface proteins among poxviruses exhibit approximately 93-98% sequence similarity based on comparison between vaccinia virus (VACV) and MPXV [6], suggesting similar roles in the entry process. Firstly, certain surface proteins located on the envelope bind to the host's extracellular glycosaminoglycans (GAGs), inducing cell adhesion [7-9]. D8L (E8L of MPXV) binds to chondroitin sulfate, while A27L (A29L of MPXV) and H3L (H3L of MPXV) bind to heparan sulfate. Additionally, A26 interacts with laminin. The receptor bound by L1R (M1R of MPXV) remains unidentified, and the viral protein that is expected to bind to the macrophage receptor with collagenous structure (MARCO) has not been identified yet. After each adhesion, A34 and B5 (B6 of MPXV) induce the rupture of the envelope membrane, exposing the inner membrane [10]. On the other hand, it has been revealed that not all of the proteins involved in cell adhesion are indispensable for infection [9]. The exposed inner membrane forms hemifusion state under lower pH conditions with a host membrane, and that caused by entry fusion complex (EFC) constructed by various viral surface proteins such as A16, A21, A28, F9, G3, G9, H2, J5, L1, and L5 [9, 11]. Subsequently, the entry of the MPXV core is completed through the membrane pore formed in the hemifusion portion (Fig. 1B). The released MPXV genomic DNA forms concentrated complex known as DNA factory immediately to undergo the stages of transcription, replication, and translation, leading to the assembly of immature virions (IV), which then become intracellular mature virions (IMV) through morphogenesis in cytoplasm (Fig. 1B) [12, 13]. Depending on the formation of a double-membrane envelope in the Golgi, the virus can escape in two forms: the intracellular enveloped virion (IEV), which is released outside the cell through exocytosis, and IMV, which can also be released without enveloping through host cell lysis (Fig. 1B). Although the surface glycoprotein, envelope composition, and receptor interaction mechanisms are not fully understood, both viral forms are infectious to neighboring host cells [11, 14].
In this review, we compiled information on the global epidemiology, diagnosis, treatment, and vaccine development status following the Mpox outbreak. In order to countermeasure Mpox outbreak, we have also summarized the content related to specific therapeutic agents and vaccine candidates for Mpox. This review will contribute to the early stages of basic research and antiviral studies related to MPXV and provides assistance in preparing for pandemic outbreaks caused by novel MPXV variants.
Epidemiology
As of May 16, 2023, 87,479 confirmed cases of Mpox and 1,095 probable cases, including 140 deaths, were reported globally by the WHO [2]. The global epidemic curve peaked in August 2022, with weekly reported cases over 7,000 (Fig. 2A). Since then, there has been a gradual decrease, and as of May 2023, approximately 100 weekly cases have been recorded. Among the confirmed cases, the United States reported the highest number (59,343), followed by Europe (25,888). The African region had a relatively low number of cases (1,592). Notably, the western Pacific region showed a gradual increase in cases since March 2023 (Fig. 2B), with approximately 40 weekly cases recorded. Australia (145), Japan (135), China (87), and South Korea (75) accounted for the majority of cases, with China and Korea experiencing an increase of cases since April 2023.
-
Fig. 2. Epidemic curves of the confirmed Mpox cases.
The presentation of the case counts is divided into overall data from Jan 1, 2022, to May 10, 2023 (A) and recent data from Mar 1, 2023, to May 10, 2023 (B). All the results were calculated and visualized based on half the value of the month.
Of all the WHO-reported types of transmission, sexual encounters were the most common (82.0%). Of all confirmed cases with available data, 96.2% were male and the median age was 34 years. Among those with known data on sexual orientation, 84.1% were categorized as men who had sex with men. In addition, 52.3% of the cases with available HIV status were HIV-positive.
Thus far, two major genotypes of MPXV have been identified. The genotype originating in Central Africa (the Congo Basin) has been classified as Clade I, while the one originating in West Africa has been designated as Clade II, which was subsequently renamed as Clade IIa after the global outbreak. Clade I has been reported to have a higher infectivity and fatality rate than Clade II (10.6%:3.6%) [15]. This was attributed to genomic deletions and fragmentations in the ORF of Clade II, which led to a decrease in virulence. In the case of Clade I, it is based on the inhibition of T-cell activation, which suppresses the expression of pro-inflammatory cytokines [1, 16]. The current world outbreak (2022–2023) was identified as Clade IIb, a subtype of the West African clade with low severity, presumably exported out of Africa and amplified by travelers who had sexual contact with men [2]. Currently, most infected people belong to the B.1 lineage of Clade IIb, and some belong to the A.2 lineage. Lineage B.1 has accumulated over 50 nucleotide changes compared to a Clade IIb MPXV isolated in 2018 [17]. Although the total fatality rate of cases counted after the outbreak was significantly lower at 0.16% (deaths/confirmed cases:140/87,479), these many nucleotide changes are likely to have influenced the activity alteration of APOBEC3 cytosine deaminase and the substantial increase in human-to-human transmission [18].
Diagnosis
The analytical methods developed for the detection of MPXV are typically conducted in biosafety level 3 facilities and are similar to the methods for other viruses. The most efficient method for sample preservation and fast analysis involves real-time polymerase chain reaction (PCR) using viral DNA isolated from lesion tissue. Since MPXV is rarely detected except during the short period of initial infection, the PCR analysis using serum samples is not recommended by the WHO [19]. Additionally, methods for detecting viral proteins and antibodies in serum are prioritized and widely utilized. Electron microscopy analysis of viruses isolated from clinical specimens and immunohistochemistry of tissue samples are also available, but they have limitations, as they require skilled techniques and specialized analytical equipment.
The diagnostic targets of MPXV are diverse. Among MPXV genes, the D6R gene of MPXV is a conserved region (> 100 nucleotides in length) that spans the entire
As an immunological method in clinical area, enzyme-linked immunosorbent assay (ELISA) which has been developed to detect patient antibodies (IgG, IgM) against Orthopoxvirus is still used to MPXV as well in the serum at five days or more than eight days after the onset of rash [27, 28]. However, the detection specificity of these antibodies is insufficient to distinguish because of the highly conserved antigen epitopes between MPXV and other poxviruses [1]. In addition, ELISA for detecting IgG and IgM has the disadvantage of being unable to distinguish between people infected with other poxviruses in the past, people who have been vaccinated, and people infected with Mpox [1, 27, 29]. Consequently, the use of this method is limited to epidemiologic studies [30].
Antiviral Therapies
Currently, no specific antiviral therapy has been approved for treating human MPXV infections. During the ongoing Mpox outbreak, supportive care and pain control are recommended as the initial treatment options for most patients. Owing to their broad-spectrum antiviral activity, several therapies (cidofovir, brincidofovir, tecovirimat, and vaccinia immunoglobulin) for smallpox infection are considered for the treatment of severe cases of Mpox and high-risk patients such as immunocompromised people, infants, pregnant or breastfeeding women, and people with skin integrity issues (Treatment Information for Healthcare Professionals from CDC [31]).
A viral replication step has been the most important antiviral target as nucleotide analogs can interfere synthesis of viral genomes. Cidofovir, an FDA-approved drug used to treat cytomegalovirus infections, inhibits DNA synthesis and interferes with replication of various DNA viruses. Owing to its broad-spectrum activity, cidofovir is considered a potential therapeutic option against other DNA viruses, including orthopoxviruses. Cidofovir remains an investigational treatment option for smallpox [32] and Mpox. Brincidofovir is a cidofovir prodrug that contains a lipid conjugate for better membrane permeability in infected cells [33]. Brincidofovir showed higher protective efficacy than cidofovir against various DNA viruses, such as orthopoxviruses [34], herpesviruses [35], and adenoviruses [36]. Recently, FDA approved brincidofovir for the treatment of smallpox. Both cidofovir and brincidfovir are nucleoside analogs that are incorporated into newly synthesized DNA strands, terminating DNA chain elongation [37].
Viral envelope is another important antiviral drug target as it plays a critical role not only in maintaining virion and protecting from diverse environment but also in viral entry. Tecovirimat, another FDA-approved drug used to treat smallpox, targets a specific orthopoxvirus membrane protein p37 (F13L of VACV), which is responsible for forming the envelope of immature intracellular virus particles [38], eventually interfering with cell-to-cell and systemic virus transmission [37, 39]. Since p37 is highly conserved in orthopoxviruses, the p37-targeting drug tecovirimat is considered a potential therapeutic option for ongoing Mpox outbreaks and is currently undergoing various clinical trials.
Each stage of the viral life cycle may be considered a potential target for the development of new antiviral drugs. Many investigational anti-orthopoxviral drugs that target key components at different stages, from viral entry to progeny release, have been identified (Table 1).
-
Table 1 . Candidate anti-orthopoxvirus agents with defined targets.
Stage of life cycle Candidate agent Mechanism of action Stage of development Ref Entry Vaccinia immune globulin Neutralizing viral particles Approved for vaccinia infection [40, 41] Early transcription Adenosine N1-oxide Nigericin
Aurintricarboxylic acidInhibiting viral early gene transcription Preclinical research [42]
[43]
[44]DNA synthesis Cidofovir Nucleoside analogs Available through IND* protocol for smallpox [37] Brincidofovir Approved for smallpox / Clinical trial phase 3 for dsDNA viruses including MPOX [45] N-Methanocarbathymidine
Aphidicolin
Cytosine arabinoside
Phosphonoacetic acid
4'-thioIDU
5-substituted deoxyuridine analogsPreclinical research [46]
[47]
[48]
[48]
[49]
[50]Small peptide aptamers Binding to virus replication complex Preclinical research [51, 52] Hydroxyurea Ribonucleotide reductase inhibitor Preclinical research [53] Late transcription IBT
Methisazone
Ethacrynic acid
Alpha-lipoic acidInhibiting viral late gene transcription Preclinical research [54, 55]
[57]
[58]
[58]Assembly Rifampicin Interfering formation of viral membrane Preclinical research [59, 60] Mitoxantrone DNA ligase inhibitor Preclinical research [61, 62] Ofloxacin
NovobiocinTopoisomerase inhibitor Preclinical research [63]
[64, 65]Maturation TTP-6171 Cysteine proteinase inhibitor Preclinical research [66] Secondary envelopment Tecovirimat VP37 inhibitor Approved for smallpox / Clinical trial phase 3 for MPOX [68] IMCBH Preventing intracellular virion wrapping Preclinical research [70, 71] Egress Imatinib Inhibiting cellular
Abl-family tyrosine kinasesPreclinical research [72-74] Terameprocol Inhibiting actin tail formation Preclinical research [75] Nitroxoline Inhibiting the signal cascade that modulates virus replication Preclinical research [76-78] *Investigational new drug (IND)
I. Entry and uncoating
• Vaccinia immune globulin [40, 41]: Neutralizing activity of antibodies blocks the interaction between virions and host receptors, inhibiting viral entry.
II. Early transcription
• Adenosine N1-oxide [42]: Incorporation of adenosine N1-oxide into early viral transcripts interferes with the translation of viral mRNA, not cellular mRNA, blocking early viral gene expression.
• Nigericin [43]: Although its precise mechanism of action remains unknown, nigericin inhibits viral replication at the early transcription stage. As a carboxylic ionophore, nigericin can be readily incorporated into biological membranes to stimulate the exchange of monovalent cations with protons. These environmental changes may affect the viral mRNA synthesis.
• Aurintricarboxylic acid [44]: Aurintricarboxylic acid inhibits the transcription of early viral genes by targeting both the cellular (blocking the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling cascade, which is essential for VACV replication) and viral factors (inhibiting the phosphatase activity of the viral enzyme H1L, which is required to initiate viral transcription).
III. DNA synthesis
• Nucleoside analogs: Nucleoside analogs mimic natural nucleosides to compete with natural nucleosides during DNA synthesis and are incorporated into newly synthesized DNA strands. Once incorporated, nucleoside analogs terminate the DNA chain elongation. Numerous nucleoside analogs have been identified as antiviral therapeutics, and several compounds have been confirmed to induce broad-spectrum anti-orthopoxviral activities (listed below).
◆ Cidofovir & brincidofovir [37, 45]
◆ N-methanocarbathymidine [46]
◆ Aphidicolin [47]
◆ Cytosine arabinoside & phosphonoacetic acid [48]
◆ 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU) [49]
◆ 5-substituted deoxyuridine analogs [50]: Viral thymidine kinase phosphorylates these modified molecules, enhancing their antiviral activities.
• Small peptide aptamers [51, 52]: Peptide aptamers targeting A20, a major component of the viral replication complex, impair viral DNA synthesis.
• Hydroxyurea [53] is a ribonucleotide reductase inhibitor that limits deoxyribonucleotide production, thereby inhibiting viral DNA synthesis.
IV. Late transcription
• Isatin-beta-thiosemicarbazone (IBT) [54, 55]: The precise mechanism of action of IBT is not completely understood. However, IBT terminates late viral gene transcripts by degrading late viral mRNAs, followed by the cessation of late viral protein production. Moreover, IBT indirectly regulates viral transcription elongation factors [56].
• Methisazone (N-methyl-isatin-/i-thiosemicarbazone, N-MeIBT,MelBT) [57]: Methisazone inhibits mRNA and protein synthesis and has antiviral activity. Methisazone has been reported to be effective in smallpox prophylaxis.
• Ethacrynic and alpha-lipoic acid [58]: Both ethacrynic and alpha-lipoic acids inhibit vaccinia late gene expression, but not viral entry, early gene expression, and viral DNA synthesis.
V. Assembly
• Rifampicin [59, 60]: Rifampicin interferes with the formation of viral membranes during the early assembly step, resulting in the inhibition of viral morphogenesis.
• Mitoxantrone [61, 62]: As a DNA ligase inhibitor, mitoxantrone blocks virion assembly, but its protective efficacy is limited in vivo.
• Ofloxacin [63]: Ofloxacin inhibits topoisomerase, which is essential for virion assembly.
• Novobiocin [64, 65]: Novobiocin is also a topoisomerase inhibitor, interfering with virion assembly.
VI. Maturation
• TTP-6171 [66]: TTP-6171 inhibits I7L core proteinase, a cysteine proteinase that processes viral structural and membrane proteins, and blocks the maturation of intracellular virions.
• Recently, a computer modeling strategy identified several potential protease inhibitors with better binding affinity than that of TTP-6171 [67]: (1) Gallicynoic Acid F (NPA002071), (2) H2-Erythro-Neopterin (NPA000530), (3) Nigcollin C (NPA029767), (4) NPA24545, and (5) Vaccinol M (NPA030378)
VII. Secondary envelopment
• Tecovirimat [68]: A VP37-targeting agent tecovirimat inhibits the formation of enveloped viral particles. Formation of this envelope is important for viral egress and the next round of infection in other cells. Accordingly, targeting VP37 with a tecovirimat prevents the viral spread to other cells in the body. Its protective efficacy against the circulating Mpox strain has been confirmed in vitro [69] and clinical trials are in progress.
• N1-isonicotinoyl-N2-3-methyl-4-chloro-benzoylhydrazine (IMCBH) [70, 71]: IMCBH is another agent that affects secondary envelope formation by preventing the wrapping of intracellular virions by Golgi membranes.
VIII. Release of progeny virus particles
• Imatinib [72-74]: As a host-targeting agent, imatinib inhibits cellular Abl family tyrosine kinases, which are required for the release of infectious progeny viral particles.
• Terameprocol [75]: Terameprocol interferes the formation of actin tails which are crucial for spreading viruses to neighboring cells.
• Host targeting drug, nitroxoline [76]: Although its mechanism of action is not known, nitroxoline inhibits the phosphoinositide 3‐kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway and stimulates the fibrosarcoma/mitogen‐activated protein kinase/extracellular signal‐regulated kinase (RAF/MEK/ERK) signaling pathway. Both signaling cascades are involved in orthopoxvirus replication [77, 78].
Vaccines
The currently available smallpox vaccine Dryvax, a live VACV that protects humans from both smallpox and Mpox, is a cloned vaccine strain [79]. However, large doses of these vaccines can cause serious side effects in high-risk individuals with acquired or congenital immune system defects [80]. Interestingly, the main mode of protection of an unattenuated smallpox vaccine against Mpox is mediated by neutralizing antibodies (Abs) [81]. Unattenuated Dryvax, modified vaccinia Ankara (MVA), and NYVAC do not protect against lethal Mpox in immunodeficient animals infected with the simian immunodeficiency virus (SIV) [82]. This was confirmed to be due to defective maturation of high-affinity protective Abs under CD4+ T cell depletion. LC16m8 [83], a VACV-mediated vaccine attenuated by passaging in mammalian primary cell, and VACΔ6 [84], a VACV-mediated vaccine intentionally modified six genes, also remain in phase 2 and 1 clinical trials respectively as a vaccine against MPXV yet (All clinical status was referenced in the ‘WHO Mpox Vaccine Tracker’ updated on April 24, 2023 [85]).
Orthopoxviruses, a family of double-stranded DNA viruses, comprise a variety of viruses, including human MPXV, variola virus, VACV, and cowpox virus [86]. Orthopoxvirus infection and immunization can confer immunity to other viruses, and as a result, two vaccines have been approved by the FDA to prevent MPXV [87]. ACAM2000 is a second-generation live VACV vaccine, and Bavarian Nordic (JYNNEOS, also known as MVA-BN, Imvamune, or Imvanex) was developed as a modified vaccinia Ankara (MVA)-based attenuated third-generation smallpox vaccine [88]. Since approval, the indication has been expanded based on the data from the MPXV challenge study conducted in non-humans (FDA's authorized “JYNNEOS” vaccine for counteracting Mpox global public health emergency). These two VACV-based smallpox vaccines cross-protect against MPXV [6]. Unfortunately, smallpox vaccines do not completely protect against circulating MPXV [89]. ACAM2000 is a highly regenerated VACV vaccine with serious side effects [90] (Table 2). In contrast, JYNNEOS, which is based on a live attenuated orthopoxvirus, is expected to have the lowest incidence of severe adverse events as a replication-deficient vaccine [90].
-
Table 2 . Approved vaccines to prevent MPXV infection.
Trade name Manufacturer Molecule type Target disease Inoculation site Dosing interval and number Injection volume Side effects JYNNEOS/ Imvanex Bavarian
Nordic ASLive
Attenuated
VaccineMonkeypox; smallpox; vaccinia virus (EU only) Subcutaneous
Intradermal2 doses administered 28 days (4 weeks) apart 0.5 ml
0.1 mlPain at the injection site, redness, itching, fever, headache, tiredness, nausea, chills, and muscle aches ACAM2000 Gaithersburg, Inc. Live virus
VaccineSmallpox Percutaneous Single dose using a bifurcated needle 0.0025 ml droplet of reconstituted vaccine (100 doses) Itching, sore arm, fever, headache, body ache, mild rash, fatigue, myocarditis, and pericarditis
Development of Novel Mpox Vaccines
Currently, mRNA- and DNA-based vaccines are being developed. Among them, one of the mRNA vaccine is the combination of mRNA-A-LNP and mRNA-B-LNP which are designed to contain two IMV-specific proteins (A29L and M1R) and two infected EEV-specific proteins (A35R and B6R) of MPXV, respectively [91]. Combinations of IMV- and EEV-specific immunogens have been studied to provide more protection than immunogens alone [92]. In a recent study, the immunogenicity of a multivalent mRNA vaccine candidate, MPXVac-097 (A29L, A35R, M1R, E8L, and B6R linked to a 2a peptide), was characterized in a mouse model [93]. MPXVac-097 elicits MPXV-specific T-cell responses and neutralizes VACV infections. As other effective mRNA vaccine candidates against MPXV, Rmix4 and Rmix6 encode each four (M1R, A29L, B6R, A35R) and six (M1R, H3L, A29L, E8L, B6R, and A35R) of MPXV antigens [94]. Freyn
Alternatively, several VACV genes and gene combinations have been tested for their immunogenicity and protective efficacy in mice using a gene gun delivery DNA vaccine. [97]. The four-gene combination DNA vaccine showed immunogenicity in non-human primates and protected 100% of the VACV-infected mice. DNA vaccines (expressing A27L, A33R, L1R, and B5R), named 4pox, have been reported to protect against fatal Mpox, rabbit pox, and VACV infections [98, 99]. Interestingly, vaccination of plasmid DNAs encoding these four antigens (L1R, A27L, A33R, and B5R) with boosting by the equivalent recombinant proteins showed much improved vaccine efficacy against MPXV [100]. In addition, Hirao
Franceschi
Conventional recombinant protein-based vaccines are currently being developed. Because live poxviral vectors express many cytokines and chemokines, it is difficult to analyze the effects of immunomodulatory approaches [103]. On the other hands, vaccination of the recombinant subunit platform comprising A33R, B5R, L1R, and A27L of VACV with Alum adjuvant and CpG was reported successful IgG generation and full protection against VACV, even in a single-targeting vaccination trials [104, 105]. This study presents a simple subunit-based vaccine with excellent potential to increase immunogenicity and protective efficacy in high-risk populations.
Synthetic vaccines have recently been proposed as new tools for controlling infectious diseases. The novel immunogens containing a mixture of synthetic peptides, which were site-specifically designed for epitopes exposed on the multiple proteins of pathogen, has been shown to be an efficient approach as vaccine development [106]. Importantly, these synthetic vaccines promote the production of antibodies that neutralize the viral proteins when administered in monkeys and were able to delay or suppress the disease development in immunized human volunteers [107]. This strategy can be reliably used for diseases caused by viruses such as MPXV by including immunogenic epitopes and appropriate immune stimulants [108]. This allows development at a relatively low cost, particularly owing to low biosafety requirements, and presents remarkable immune diversity [109].
-
Fig. 3. The characteristics of vaccine candidates against Mpox.
Summary
Currently available therapeutics and vaccines for smallpox are cross-reactive to MPXV [6, 31, 32]. However, there are limitations to the therapeutic efficacy of different treatment and vaccine products against Mpox; thus, the development of specific therapeutics and vaccines against Mpox is still required [89]. One of them is that VACV exist in two forms (IMV and EEV) and infect cells by different mechanisms [11]. Thus, it is highly recommended to develop antiviral drugs targeting the replication steps of the viral life cycle, which can inhibit viral propagation regardless of viral entry. For example, tecovirimat does not affect the intracellular form of the virus and can only inhibit the formation of enveloped virions because it targets the p37 membrane protein of VACV, which only exists in EEV [68, 110]. In contrast, cidofovir and brincidofovir block orthopoxvirus DNA polymerase-mediated DNA synthesis, inhibiting viral replication [37, 45].
As for the vaccine development, multivalent vaccines created by the addition of MPXV immunogens to previously developed vaccine components are key strategies for the development of Mpox vaccines. Since two forms of VACV (i.e., IMV and EEV) can enter cells by different mechanisms, the composition of immunogens may affect their neutralizing activity. Indeed, despite the high immunogenicity, sera from animals immunized with the EEV proteins did not show neutralizing activity against IMV. More importantly, protective efficacy was better in animals immunized with combinations of IMV and EEV proteins than in animals immunized with individual IMV or EEV proteins [111]. Hence, this should be considered in the development of multivalent Mpox vaccines.
Another key strategy for improving vaccines is to strengthen previously developed vaccines to cover more diverse strains of orthopoxviruses, including Mpox. This is possible by boosting cellular immune responses. Since Mpox has highly similar amino acid sequences to vaccinia viruses, T cell-mediated cellular responses induced by VACV vaccines are cross-reactive to Mpox [112]. Optimization of adjuvants or the addition of peptides conserved between the VACV and Mpox can increase T cell-mediated cellular responses, allowing vaccines to target a broad range of viruses.
Together with the development of rapid diagnostics specific for Mpox, improvements in the effectiveness of therapeutics and vaccines against Mpox will prevent future Mpox outbreaks.
Acknowledgments
This work was supported in part by the Korea Research Institute of Chemical Technology (KRICT) (Project No. KK2333-20) and the National Research Foundation of Korea (grant number NRF-2021M3E5E3080540).
Conflict of Interest
The authors have no financial conflicts of interest to declare.
References
- McCollum AM, Damon IK. 2013. Human Monkeypox.
Clin. Infect. Dis. 58 : 260-267. - WHO. 2023. 2022-23 Mpox outbreak: Global Trends. Available from https://worldhealthorg.shinyapps.io/mpx_global/. Accessed on May 16, 2023.
- Chadha J, Khullar L, Gulati P, Chhibber S, Harjai K. 2022. Insights into the monkeypox virus: Making of another pandemic within the pandemic?
Environ. Microbiol. 24 : 4547-4560. - 2023. Mpox (Monkeypox): Causes, Symptoms, Treatment & Prevention. Available from https://my.clevelandclinic.org/health/diseases/22371-monkeypox#symptoms-and-causes. Accessed on April 25, 2023.
- 2022. Investigation into monkeypox outbreak in England: technical briefing 1. Available from https://www.gov.uk/government/publications/monkeypox-outbreak-technical-briefings/investigation-into-monkeypox-outbreak-in-england-technical-briefing-1#sources-and-acknowledgments. Accessed on Sep. 23, 2023.
- Ahmed SF, Sohail MS, Quadeer AA, McKay MR. 2022. Vaccinia-virus-based vaccines are expected to elicit highly cross-reactive immunity to the 2022 monkeypox virus.
Viruses 14 : 1960. - Louten J, Reynolds N. 2016. Essential human virology, pp. 273-290. Ed. Academic Press is an imprint of Elsevier, London, UK.
- Shi D, He P, Song Y, Cheng S, Linhardt RJ, Dordick JS,
et al . 2022. Kinetic and structural aspects of glycosaminoglycan - Monkeypox virus protein A29 interactions using surface plasmon resonance.Molecules 27 : 5898. - Moss B. 2016. Membrane fusion during poxvirus entry.
Semin. Cell. Dev. Biol. 60 : 89-96. - Roberts KL, Breiman A, Carter GC, Ewles HA, Hollinshead M, Law M,
et al . 2009. Acidic residues in the membrane-proximal stalk region of vaccinia virus protein B5 are required for glycosaminoglycan-mediated disruption of the extracellular enveloped virus outer membrane.J. Gen. Virol. 90 : 1582-1591. - Gray RDM, Albrecht D, Beerli C, Huttunen M, Cohen GH, White IJ,
et al . 2019. Nanoscale polarization of the entry fusion complex of vaccinia virus drives efficient fusion.Nat. Microbiol. 4 : 1636-1644. - Moss B. 2013. Poxvirus DNA replication.
Cold Spring Harb. Perspect. Biol. 5 : a010199. - Katsafanas GC, Moss B. 2007. Colocalization of transcription and translation within cytoplasmic poxvirus factories coordinates viral expression and subjugates host functions.
Cell Host Microbe. 2 : 221-228. - Li H, Huang QZ, Zhang H, Liu ZX, Chen XH, Ye LL,
et al . 2023. The land-scape of immune response to monkeypox virus.EBioMedicine 87 : 104424. - Liang C, Qian J, Liu L. 2022. Biological characteristics, biosafety prevention and control strategies for the 2022 multi-country outbreak of monkeypox.
Biosaf. Health 04 : 376-385. - Reynolds MG, Damon IK. 2012. Outbreaks of human monkeypox after cessation of smallpox vaccination.
Trends Microbiol. 20 : 80-87. - Isidro J, Borges V, Pinto M, Sobral D, Santos JD, Nunes A,
et al . 2022. Phylogenomic characterization and signs of microevolution in the 2022 multi-country outbreak of monkeypox virus.Nat. Med. 28 : 1569-1572. - Alcami A. 2023. Pathogenesis of the circulating mpox virus and its adaptation to humans.
Proc. Natl. Acad. Sci. USA 120 : e2301662120. - WHO. 2022. Laboratory testing for the monkeypox virus: Interim guidance. Available from https://www.who.int/publications/i/item/WHO-MPX-laboratory-2022.1. Accessed on 05.23, 2022.
- Luciani L, Inchauste L, Ferraris O, Charrel R, Nougairede A, Piorkowski G,
et al . 2021. A novel and sensitive real-time PCR system for universal detection of poxviruses.Sci. Rep. 11 : 1798. - Orba Y, Sasaki M, Yamaguchi H, Ishii A, Thomas Y, Ogawa H,
et al . 2015. Orthopoxvirus infection among wildlife in Zambia.J. Gen. Virol. 96 : 390-394. - Li Y, Olson VA, Laue T, Laker MT, Damon IK. 2006. Detection of monkeypox virus with real-time PCR assays.
J. Clin. Virol. 36 : 194-203. - Shchelkunov SN, Shcherbakov DN, Maksyutov RA, Gavrilova EV. 2011. Species-specific identification of variola, monkeypox, cowpox, and vaccinia viruses by multiplex real-time PCR assay.
J. Virol. Methods 175 : 163-169. - Kulesh DA, Loveless BM, Norwood D, Garrison J, Whitehouse CA, Hartmann C,
et al . 2004. Monkeypox virus detection in rodents using real-time 3'-minor groove binder TaqMan assays on the Roche LightCycler.Lab. Invest. 84 : 1200-1208. - CDC. 2023. Mpox Generic PCR Protocol. Available from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on March 7, 2023.
- Li Y, Zhao H, Wilkins K, Hughes C, Damon IK. 2010. Real-time PCR assays for the specific detection of monkeypox virus West African and Congo Basin strain DNA.
J. Virol. Methods 169 : 223-227. - WHO. 2022. Surveillance, case investigation and contact tracing for mpox (monkeypox): interim guidance. Available from https://www.who.int/publications/i/item/WHO-MPX-Surveillance-2022.4. Accessed on 12.22, 2022.
- Karem KL, Reynolds M, Braden Z, Lou G, Bernard N, Patton J,
et al . 2005. characterization of acute-phase humoral immunity to monkeypox: use of immunoglobulin M enzyme-linked immunosorbent assay for detection of monkeypox infection during the 2003 North American outbreak.Clin. Diagn. Lab. Immunol. 12 : 867-872. - Karem KL, Reynolds M, Braden Z, Lou G, Bernard N, Patton J,
et al . 2005. Characterization of acute-phase humoral immunity to Monkeypox: Use of immunoglobulin M enzyme-linked immunosorbent assay for detection of Monkeypox infection during the 2003 North American outbreak.Clin. Diagn. Lab. Immunol. 12 : 867-872. - Alakunle E, Moens U, Nchinda G, Okeke MI. 2020. Monkeypox virus in Nigeria: Infection biology, epidemiology, and evolution.
Viruses 12 : 1257. - CDC. 2023. Treatment Information for Healthcare Professionals. Available from https://www.cdc.gov/poxvirus/mpox/clinicians/treatment.html. Accessed vailable from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on April 21, 2023.
- FDA. 2023. Smallpox Preparedness and Response Updates from FDA. Available from https://www.fda.gov/emergencypreparedness-and-response/mcm-issues/smallpox-preparedness-and-response-updates-fda. Accessed vailable from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on May 3, 2023.
- Hostetler KY. 2009. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art.
Antiviral Res. 82 : A84-98. - Kern ER, Hartline C, Harden E, Keith K, Rodriguez N, Beadle JR,
et al . 2002. Enhanced inhibition of orthopoxvirus replication in vitro by alkoxyalkyl esters of cidofovir and cyclic cidofovir.Antimicrob. Agents Chemother. 46 : 991-995. - Beadle JR, Hartline C, Aldern KA, Rodriguez N, Harden E, Kern ER,
et al . 2002. Alkoxyalkyl esters of cidofovir and cyclic cidofovir exhibit multiple-log enhancement of antiviral activity against cytomegalovirus and herpesvirus replication in vitro.Antimicrob. Agents Chemother. 46 : 2381-2386. - Hartline CB, Gustin KM, Wan WB, Ciesla SL, Beadle JR, Hostetler KY,
et al . 2005. Ether lipid-ester prodrugs of acyclic nucleoside phosphonates: activity against adenovirus replication in vitro.J. Infect. Dis. 191 : 396-399. - Siegrist EA, Sassine J. 2023. Antivirals with activity against Mpox: A clinically oriented review.
Clin. Infect. Dis. 76 : 155-164. - Yang G, Pevear DC, Davies MH, Collett MS, Bailey T, Rippen S,
et al . 2005. An orally bioavailable antipoxvirus compound (ST-246) inhibits extracellular virus formation and protects mice from lethal orthopoxvirus challenge.J. Virol. 79 : 13139-13149. - Grosenbach DW, Jordan R, Hruby DE. 2011. Development of the small-molecule antiviral ST-246 as a smallpox therapeutic.
Future Virol. 6 : 653-671. - Bell E, Shamim M, Whitbeck JC, Sfyroera G, Lambris JD, Isaacs SN. 2004. Antibodies against the extracellular enveloped virus B5R protein are mainly responsible for the EEV neutralizing capacity of vaccinia immune globulin.
Virology 325 : 425-431. - Thet AK, Kelly PJ, Kasule SN, Shah AK, Chawala A, Latif A,
et al . 2023. The use of vaccinia immune globulin in the treatment of severe Mpox. virus infection in human immunodeficiency virus/AIDS.Clin. Infect. Dis. 76 : 1671-1673. - Kane EM, Shuman S. 1995. Adenosine N1-oxide inhibits vaccinia virus replication by blocking translation of viral early mRNAs.
J. Virol. 69 : 6352-6358. - Myskiw C, Piper J, Huzarewich R, Booth TF, Cao J, He R. 2010. Nigericin is a potent inhibitor of the early stage of vaccinia virus replication.
Antiviral Res. 88 : 304-310. - Myskiw C, Deschambault Y, Jefferies K, He R, Cao J. 2007. Aurintricarboxylic acid inhibits the early stage of vaccinia virus replication by targeting both cellular and viral factors.
J. Virol. 81 : 3027-3032. - Rizk JG, Lippi G, Henry BM, Forthal DN, Rizk Y. 2022. Prevention and treatment of Monkeypox.
Drugs 82 : 957-963. - Prichard MN, Keith KA, Quenelle DC, Kern ER. 2006. Activity and mechanism of action of
N -methanocarbathymidine against herpesvirus and orthopoxvirus infections.Antimicrob. Agents Chemother. 50 : 1336-1341. - Pedrali-Noy G, Spadari S. 1980. Mechanism of inhibition of herpes simplex virus and vaccinia virus DNA polymerases by aphidicolin, a highly specific inhibitor of DNA replication in eucaryotes.
J. Virol. 36 : 457-464. - Taddie JA, Traktman P. 1993. Genetic characterization of the vaccinia virus DNA polymerase: cytosine arabinoside resistance requires a variable lesion conferring phosphonoacetate resistance in conjunction with an invariant mutation localized to the 3'-5' exonuclease domain.
J. Virol. 67 : 4323-4336. - Kern ER, Prichard MN, Quenelle DC, Keith KA, Tiwari KN, Maddry JA,
et al . 2009. Activities of certain 5-substituted 4'-thiopyrimidine nucleosides against orthopoxvirus infections.Antimicrob. Agents Chemother. 53 : 572-579. - Smee DF, Humphreys DE, Hurst BL, Barnard DL. 2008. Antiviral activities and phosphorylation of 5-halo-2'-deoxyuridines and Nmethanocarbathymidine in cells infected with vaccinia virus.
Antivir. Chem. Chemother. 19 : 15-24. - Saccucci L, Crance JM, Colas P, Bickle M, Garin D, Iseni F. 2009. Inhibition of vaccinia virus replication by peptide aptamers.
Antiviral Res. 82 : 134-140. - Flusin O, Saccucci L, Contesto-Richefeu C, Hamdi A, Bardou C, Poyot T,
et al . 2012. A small molecule screen in yeast identifies inhibitors targeting protein-protein interactions within the vaccinia virus replication complex.Antiviral Res. 96 : 187-195. - Slabaugh MB, Mathews CK. 1986. Hydroxyurea-resistant vaccinia virus: overproduction of ribonucleotide reductase.
J. Virol. 60 : 506-514. - Condit RC, Easterly R, Pacha RF, Fathi Z, Meis RJ. 1991. A vaccinia virus isatin-beta-thiosemicarbazone resistance mutation maps in the viral gene encoding the 132-kDa subunit of RNA polymerase.
Virology 185 : 857-861. - Meis RJ, Condit RC. 1991. Genetic and molecular biological characterization of a vaccinia virus gene which renders the virus dependent on isatin-beta-thiosemicarbazone (IBT).
Virology 182 : 442-454. - Prins C, Cresawn SG, Condit RC. 2004. An isatin-beta-thiosemicarbazone-resistant vaccinia virus containing a mutation in the second largest subunit of the viral RNA polymerase is defective in transcription elongation.
J. Biol. Chem. 279 : 44858-44871. - Weiss MM, Weiss PD, Mathisen G, Guze P. 2004. Rethinking smallpox.
Clin. Infect. Dis. 39 : 1668-1673. - Spisakova M, Cizek Z, Melkova Z. 2009. Ethacrynic and alpha-lipoic acids inhibit vaccinia virus late gene expression.
Antiviral Res. 81 : 156-165. - Charity JC, Katz E, Moss B. 2007. Amino acid substitutions at multiple sites within the vaccinia virus D13 scaffold protein confer resistance to rifampicin.
Virology 359 : 227-232. - Baldick CJ Jr., Moss B. 1987. Resistance of vaccinia virus to rifampicin conferred by a single nucleotide substitution near the predicted NH2 terminus of a gene encoding an Mr 62,000 polypeptide.
Virology 156 : 138-145. - Deng L, Dai P, Ciro A, Smee DF, Djaballah H, Shuman S. 2007. Identification of novel antipoxviral agents: mitoxantrone inhibits vaccinia virus replication by blocking virion assembly.
J. Virol. 81 : 13392-13402. - Altmann SE, Smith AL, Dyall J, Johnson RF, Dodd LE, Jahrling PB,
et al . 2012. Inhibition of cowpox virus and monkeypox virus infection by mitoxantrone.Antiviral Res. 93 : 305-308. - Ikeda S, Yazawa M, Nishimura C. 1987. Antiviral activity and inhibition of topoisomerase by ofloxacin, a new quinolone derivative.
Antiviral Res. 8 : 103-113. - Sekiguchi J, Stivers JT, Mildvan AS, Shuman S. 1996. Mechanism of inhibition of vaccinia DNA topoisomerase by novobiocin and coumermycin.
J. Biol. Chem. 271 : 2313-2322. - Sekiguchi J, Shuman S. 1997. Novobiocin inhibits vaccinia virus replication by blocking virus assembly.
Virology 235 : 129-137. - Byrd CM, Bolken TC, Mjalli AM, Arimilli MN, Andrews RC, Rothlein R,
et al . 2004. New class of orthopoxvirus antiviral drugs that block viral maturation.J. Virol. 78 : 12147-12156. - Dubey A, Alawi MM, Alandijany TA, Alsaady IM, Altwaim SA, Sahoo AK,
et al . 2023. Exploration of microbially derived natural compounds against Monkeypox virus as viral core cysteine proteinase inhibitors.Viruses 15 : 251. - Prichard MN, Kern ER. 2012. Orthopoxvirus targets for the development of new antiviral agents.
Antiviral Res. 94 : 111-125. - Frenois-Veyrat G, Gallardo F, Gorge O, Marcheteau E, Ferraris O, Baidaliuk A,
et al . 2022. Tecovirimat is effective against human monkeypox virus in vitro at nanomolar concentrations.Nat. Microbiol. 7 : 1951-1955. - Payne LG, Kristenson K. 1979. Mechanism of vaccinia virus release and its specific inhibition by N1-isonicotinoyl-N2-3-methyl-4-chlorobenzoylhydrazine.
J. Virol. 32 : 614-622. - Schmutz C, Payne LG, Gubser J, Wittek R. 1991. A mutation in the gene encoding the vaccinia virus 37,000-M(r) protein confers resistance to an inhibitor of virus envelopment and release.
J. Virol. 65 : 3435-3442. - Reeves PM, Smith SK, Olson VA, Thorne SH, Bornmann W, Damon IK,
et al . 2011. Variola and monkeypox viruses utilize conserved mechanisms of virion motility and release that depend on abl and SRC family tyrosine kinases.J. Virol. 85 : 21-31. - Newsome TP, Weisswange I, Frischknecht F, Way M. 2006. Abl collaborates with Src family kinases to stimulate actin-based motility of vaccinia virus.
Cell. Microbiol. 8 : 233-241. - Reeves PM, Bommarius B, Lebeis S, McNulty S, Christensen J, Swimm A,
et al . 2005. Disabling poxvirus pathogenesis by inhibition of Abl-family tyrosine kinases.Nat. Med. 11 : 731-739. - Pollara JJ, Laster SM, Petty IT. 2010. Inhibition of poxvirus growth by Terameprocol, a methylated derivative of nordihydroguaiaretic acid.
Antiviral Res. 88 : 287-295. - Bojkova D, Zoller N, Tietgen M, Steinhorst K, Bechtel M, Rothenburger T,
et al . 2023. Repurposing of the antibiotic nitroxoline for the treatment of mpox.J. Med. Virol. 95 : e28652. - Kindrachuk J, Arsenault R, Kusalik A, Kindrachuk KN, Trost B, Napper S,
et al . 2012. Systems kinomics demonstrates Congo Basin monkeypox virus infection selectively modulates host cell signaling responses as compared to West African monkeypox virus.Mol. Cell. Proteomics 11 : M111015701. - Beerli C, Yakimovich A, Kilcher S, Reynoso GV, Flaschner G, Muller DJ,
et al . 2019. Vaccinia virus hijacks EGFR signalling to enhance virus spread through rapid and directed infected cell motility.Nat. Microbiol. 4 : 216-225. - Hammarlund E, Lewis MW, Carter SV, Amanna I, Hansen SG, Strelow LI,
et al . 2005. Multiple diagnostic techniques identify previously vaccinated individuals with protective immunity against monkeypox.Nat. Med. 11 : 1005-1011. - Kemper AR, Davis MM, Freed GL. 2002. Expected adverse events in a mass smallpox vaccination campaign.
Eff. Clin. Pract. 5 : 84-90. - Edghill-Smith Y, Golding H, Manischewitz J, King LR, Scott D, Bray M,
et al . 2005. Smallpox vaccine-induced antibodies are necessary and sufficient for protection against monkeypox virus.Nat. Med. 11 : 740-747. - Edghill-Smith Y, Bray M, Whitehouse CA, Miller D, Mucker E, Manischewitz J,
et al . 2005. Smallpox vaccine does not protect macaques with AIDS from a lethal monkeypox virus challenge.J. Infect. Dis. 191 : 372-381. - Kenner J, Cameron F, Empig C, Jobes DV, Gurwith M. 2006. LC16m8: an attenuated smallpox vaccine.
Vaccine 24 : 7009-7022. - Maksyutov RA, Yakubitskiy SN, Kolosova IV, Tregubchak TV, Shvalov AN, Gavrilova EV,
et al . 2022. Genome stability of the vaccine strain VACDelta6.Vavilovskii Zhurnal Genet Selektsii. 26 : 394-401. - WHO. 2023. Mpox Vaccine Tracker: List of vaccine candidates in research & development. Available from https://www.who.int/publications/m/item/mpox-vaccine-tracker---list-of-vaccine-candidates-in-research---development. Accessed on April 23, 2023.
- Lum FM, Torres-Ruesta A, Tay MZ, Lin RTP, Lye DC, Renia L,
et al . 2022. Monkeypox: disease epidemiology, host immunity and clinical interventions.Nat. Rev. Immunol. 22 : 597-613. - Petersen BW, Harms TJ, Reynolds MG, Harrison LH. 2016. 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.
MMWR Morb. Mortal. Wkly. Rep. 65 : 257-262. - Pittman PR, Hahn M, Lee HS, Koca C, Samy N, Schmidt D,
et al . 2019. Phase 3 efficacy trial of modified vaccinia ankara as a vaccine against smallpox.N. Engl. J. Med. 381 : 1897-1908. - Thornhill JP, Barkati S, Walmsley S, Rockstroh J, Antinori A, Harrison LB,
et al . 2022. Monkeypox Virus Infection in Humans across 16 Countries - April-June 2022.N. Engl. J. Med. 387 : 679-691. - Rao AK, Petersen BW, Whitehill F, Razeq JH, Isaacs SN, Merchlinsky MJ,
et al . 2022. Use of JYNNEOS (Smallpox and Monkeypox Vaccine, Live, Nonreplicating) for Preexposure Vaccination of Persons at Risk for Occupational Exposure toOrthopoxviruses : Recommendations of the Advisory Committee on Immunization Practices - United States, 2022.MMWR Morb. Mortal. Wkly. Rep. 71 : 734-742. - Sang Y, Zhang Z, Liu F, Lu H, Yu C, Sun H,
et al . 2023. Monkeypox virus quadrivalent mRNA vaccine induces immune response and protects against vaccinia virus.Signal. Transduct. Target. Ther. 8 : 172. - Hooper JW, Custer DM, Schmaljohn CS, Schmaljohn AL. 2000. DNA vaccination with vaccinia virus L1R and A33R genes protects mice against a lethal poxvirus challenge.
Virology 266 : 329-339. - Fang Z, Monteiro VS, Renauer PA, Shang X, Suzuki K, Ling X,
et al . 2023. Polyvalent mRNA vaccination elicited potent immune response to monkeypox virus surface antigens.Cell. Res. 33 : 407-410. - Zeng J, Li Y, Jiang L, Luo L, Wang Y, Wang H,
et al . 2023. Mpox multi-antigen mRNA vaccine candidates by a simplified manufacturing strategy afford efficient protection against lethal orthopoxvirus challenge.Emerg. Microbes. Infect. 12 : 2204151. - Freyn AW, Atyeo C, Earl PL, Americo JL, Chuang G-Y, Natarajan H,
et al . 2022. A monkeypox mRNA-lipid nanoparticle vaccine targeting virus binding, entry, and transmission drives protection against lethal orthopoxviral challenge.bioRxiv. : 2022.2012.2017.520886. - Hou F, Zhang Y, Liu X, Murad Y, Xu J, Yu Z,
et al . 2022. Novel mRNA vaccines encoding Monkeypox virus M1R and A35R protect mice from a lethal virus challenge.bioRxiv. : 2022.2011.2019.517190. - Hooper JW, Thompson E, Wilhelmsen C, Zimmerman M, Ichou MA, Steffen SE,
et al . 2004. Smallpox DNA vaccine protects nonhuman primates against lethal monkeypox.J. Virol. 78 : 4433-4443. - Hooper JW, Custer DM, Thompson E. 2003. Four-gene-combination DNA vaccine protects mice against a lethal vaccinia virus challenge and elicits appropriate antibody responses in nonhuman primates.
Virology 306 : 181-195. - Mucker EM, Golden JW, Hammerbeck CD, Kishimori JM, Royals M, Joselyn MD,
et al . 2022. A nucleic acid-based orthopoxvirus vaccine targeting the vaccinia virus L1, A27, B5, and A33 proteins protects rabbits against lethal rabbitpox virus aerosol challenge.J. Virol. 96 : e0150421. - Heraud JM, Edghill-Smith Y, Ayala V, Kalisz I, Parrino J, Kalyanaraman VS,
et al . 2006. Subunit recombinant vaccine protects against monkeypox.J. Immunol. 177 : 2552-2564. - Hirao LA, Draghia-Akli R, Prigge JT, Yang M, Satishchandran A, Wu L,
et al . 2011. Multivalent smallpox DNA vaccine delivered by intradermal electroporation drives protective immunity in nonhuman primates against lethal monkeypox challenge.J. Infect. Dis. 203 : 95-102. - Franceschi V, Parker S, Jacca S, Crump RW, Doronin K, Hembrador E,
et al . 2015. BoHV-4-Based vector single heterologous antigen delivery protects STAT1(-/-) Mice from Monkeypoxvirus lethal challenge.PLoS Negl. Trop. Dis. 9 : e0003850. - Hernaez B, Alcami A. 2018. New insights into the immunomodulatory properties of poxvirus cytokine decoy receptors at the cell surface
7 : F1000Res. - Buchman GW, Cohen ME, Xiao Y, Richardson-Harman N, Silvera P, DeTolla LJ,
et al . 2010. A protein-based smallpox vaccine protects non-human primates from a lethal monkeypox virus challenge.Vaccine 28 : 6627-6636. - Xiao Y, Zeng Y, Schante C, Joshi SB, Buchman GW, Volkin DB,
et al . 2020. Short-term and longer-term protective immune responses generated by subunit vaccination with smallpox A33, B5, L1 or A27 proteins adjuvanted with aluminum hydroxide and CpG in mice challenged with vaccinia virus.Vaccine 38 : 6007-6018. - Patarroyo ME, Amador R, Clavijo P, Moreno A, Guzman F, Romero P,
et al . 1988. A synthetic vaccine protects humans against challenge with asexual blood stages of Plasmodium falciparum malaria.Nature 332 : 158-161. - Lozano JM, Rodriguez Parra Z, Hernandez-Martinez S, Yasnot-Acosta MF, Rojas AP, Marin-Waldo LS,
et al . 2021. The search of a Malaria vaccine: The time for modified immuno-potentiating probes.Vaccines (Basel) 9 : 115. - Lozano JM, Muller S. 2023. Monkeypox: potential vaccine development strategies.
Trends Pharmacol. Sci. 44 : 15-19. - Bonam SR, Partidos CD, Halmuthur SKM, Muller S. 2017. An overview of novel adjuvants designed for improving vaccine efficacy.
Trends Pharmacol. Sci. 38 : 771-793. - Blasco R, Moss B. 1991. Extracellular vaccinia virus formation and cell-to-cell virus transmission are prevented by deletion of the gene encoding the 37,000-Dalton outer envelope protein.
J. Virol. 65 : 5910-5920. - Fogg C, Lustig S, Whitbeck JC, Eisenberg RJ, Cohen GH, Moss B. 2004. Protective immunity to vaccinia virus induced by vaccination with multiple recombinant outer membrane proteins of intracellular and extracellular virions.
J. Virol. 78 : 10230-10237. - Ando J, Ngo MC, Ando M, Leen A, Rooney CM. 2020. Identification of protective T-cell antigens for smallpox vaccines.
Cytotherapy 22 : 642-652.
Related articles in JMB
Article
Minireview
J. Microbiol. Biotechnol. 2023; 33(8): 981-991
Published online August 28, 2023 https://doi.org/10.4014/jmb.2306.06033
Copyright © The Korean Society for Microbiology and Biotechnology.
Current Status of Epidemiology, Diagnosis, Therapeutics, and Vaccines for the Re-Emerging Human Monkeypox Virus
Wooseong Lee, Yu-Jin Kim, Su Jin Lee, Dae-Gyun Ahn*, and Seong-Jun Kim*
Department of Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
Correspondence to:Dae-Gyun Ahn, dgahn@krict.re.kr
Seong-Jun Kim, sekim@krict.re.kr
Abstract
Monkeypox (Mpox) virus, a member of the Poxviridae family, causes a severe illness similar to smallpox, which is characterized by symptoms such as high fever, rash, and pustules. Human-to-human transmission cases have been reported but remained low since the first recorded case of human infection occurred in the Congo in 1970. Recently, Mpox has re-emerged, leading to an alarming surge in infections worldwide since 2022, originating in the United Kingdom. Consequently, the World Health Organization (WHO) officially declared the '2022–23 Mpox outbreak'. Currently, no specific therapy or vaccine is available for Mpox. Therefore, patients infected with Mpox are treated using conventional therapies developed for smallpox. However, the vaccines developed for smallpox have demonstrated only partial efficacy against Mpox, allowing viral transmission among humans. In this review, we discuss the current epidemiology of the ongoing Mpox outbreak and provide an update on the progress made in diagnosis, treatment, and development of vaccines for Mpox.
Keywords: Human monkeypox virus, epidemiology, diagnosis, therapeutics, vaccine
Introduction
Monkeypox (Mpox), a virus-borne infectious disease that recently re-emerged worldwide, has raised concerns. It exhibits symptoms similar to those of smallpox, such as high fever, generalized body aches, and the development of distinctive rashes and pustules on the skin that last a few weeks [1]. It is caused by infection with Mpox virus (MPXV), which belongs to the
MPXV is a large virus (200–250 nm in diameter) with a lipid envelope and a linear double-stranded DNA genome of approximately 197 kbp. The viral genome contains about 6.4 kbp inverted terminal repeats (ITR) and covalently closed hairpin ends. Genetic diversity at both ends affects the virulence, pathogenicity, and fatality of
The MPXV genome contains approximately 190 ORFs. However, the roles of many proteins, excluding certain key viral proteins involved in viral replication and assembly, are not yet clearly understood, although they are thought to be involved in host tropism and immunomodulation. The genome has a low GC content of approximately 33% and is characterized by the presence of numerous local tandem repeats, including the ITR. These features make it challenging to achieve a consistent reconstruction during gene replication and virion assembly [5].
MPXV is composed of several constituents (genomic DNA, nucleocapsid, core, lateral body, inner membrane, envelope, and surface proteins) (Fig. 1A). The core is dumbbell-shaped and is surrounded by the core membrane, palisade layer, and lateral bodies.
-
Figure 1. An overview of MPXV structure and lifecycle.
(A) The virion mainly consists of DNA genome, nucleocapsids, dumbbell-shaped core, lateral bodies, inner membrane, envelope, and surface proteins. There are at least 25 species of surface proteins, which are distributed on envelope and/or inner membrane, respectively. (B) The attachment and membrane fusion of virions are dependent on the interaction among viral surface proteins, extracellular GAGs, and matrix proteins of the host cell. Uncoated genomic DNA immediately forms concentrated complex known as DNA factory to undergo the stages of transcription, replication, and translation, leading to the assembly of IV. Subsequently, morphogenesis occurs in the cytoplasm to shape IV into IMV. EVs with envelope formed in the Golgi complex are released from the cells through exocytosis, and IMVs without envelope can also be released from the cells through cell lysis. GAG, glycosaminoglycan; IV, immature virion; IMV, intracellular mature virion; IEV, intracellular enveloped virion; EEV, extracellular enveloped virion.
MPXV entry occurs in a manner similar to the distinctive entry mechanism of other poxviruses; however, the detailed mechanism has not yet been fully elucidated. Herein, we briefly describe the basic entry process of vaccinia virus (VACV), which is a well-known poxvirus relatively, and the viral surface proteins involved, as well as the host receptors. It has been known that poxvirus has over 8 surface proteins on the envelope and over 20 surface proteins on the inner membrane. Adhesion to cellular receptors, membrane fusion, and inter-host transmission occur through parallel interactions between these surface proteins and their respective host receptors. Fortunately, surface proteins among poxviruses exhibit approximately 93-98% sequence similarity based on comparison between vaccinia virus (VACV) and MPXV [6], suggesting similar roles in the entry process. Firstly, certain surface proteins located on the envelope bind to the host's extracellular glycosaminoglycans (GAGs), inducing cell adhesion [7-9]. D8L (E8L of MPXV) binds to chondroitin sulfate, while A27L (A29L of MPXV) and H3L (H3L of MPXV) bind to heparan sulfate. Additionally, A26 interacts with laminin. The receptor bound by L1R (M1R of MPXV) remains unidentified, and the viral protein that is expected to bind to the macrophage receptor with collagenous structure (MARCO) has not been identified yet. After each adhesion, A34 and B5 (B6 of MPXV) induce the rupture of the envelope membrane, exposing the inner membrane [10]. On the other hand, it has been revealed that not all of the proteins involved in cell adhesion are indispensable for infection [9]. The exposed inner membrane forms hemifusion state under lower pH conditions with a host membrane, and that caused by entry fusion complex (EFC) constructed by various viral surface proteins such as A16, A21, A28, F9, G3, G9, H2, J5, L1, and L5 [9, 11]. Subsequently, the entry of the MPXV core is completed through the membrane pore formed in the hemifusion portion (Fig. 1B). The released MPXV genomic DNA forms concentrated complex known as DNA factory immediately to undergo the stages of transcription, replication, and translation, leading to the assembly of immature virions (IV), which then become intracellular mature virions (IMV) through morphogenesis in cytoplasm (Fig. 1B) [12, 13]. Depending on the formation of a double-membrane envelope in the Golgi, the virus can escape in two forms: the intracellular enveloped virion (IEV), which is released outside the cell through exocytosis, and IMV, which can also be released without enveloping through host cell lysis (Fig. 1B). Although the surface glycoprotein, envelope composition, and receptor interaction mechanisms are not fully understood, both viral forms are infectious to neighboring host cells [11, 14].
In this review, we compiled information on the global epidemiology, diagnosis, treatment, and vaccine development status following the Mpox outbreak. In order to countermeasure Mpox outbreak, we have also summarized the content related to specific therapeutic agents and vaccine candidates for Mpox. This review will contribute to the early stages of basic research and antiviral studies related to MPXV and provides assistance in preparing for pandemic outbreaks caused by novel MPXV variants.
Epidemiology
As of May 16, 2023, 87,479 confirmed cases of Mpox and 1,095 probable cases, including 140 deaths, were reported globally by the WHO [2]. The global epidemic curve peaked in August 2022, with weekly reported cases over 7,000 (Fig. 2A). Since then, there has been a gradual decrease, and as of May 2023, approximately 100 weekly cases have been recorded. Among the confirmed cases, the United States reported the highest number (59,343), followed by Europe (25,888). The African region had a relatively low number of cases (1,592). Notably, the western Pacific region showed a gradual increase in cases since March 2023 (Fig. 2B), with approximately 40 weekly cases recorded. Australia (145), Japan (135), China (87), and South Korea (75) accounted for the majority of cases, with China and Korea experiencing an increase of cases since April 2023.
-
Figure 2. Epidemic curves of the confirmed Mpox cases.
The presentation of the case counts is divided into overall data from Jan 1, 2022, to May 10, 2023 (A) and recent data from Mar 1, 2023, to May 10, 2023 (B). All the results were calculated and visualized based on half the value of the month.
Of all the WHO-reported types of transmission, sexual encounters were the most common (82.0%). Of all confirmed cases with available data, 96.2% were male and the median age was 34 years. Among those with known data on sexual orientation, 84.1% were categorized as men who had sex with men. In addition, 52.3% of the cases with available HIV status were HIV-positive.
Thus far, two major genotypes of MPXV have been identified. The genotype originating in Central Africa (the Congo Basin) has been classified as Clade I, while the one originating in West Africa has been designated as Clade II, which was subsequently renamed as Clade IIa after the global outbreak. Clade I has been reported to have a higher infectivity and fatality rate than Clade II (10.6%:3.6%) [15]. This was attributed to genomic deletions and fragmentations in the ORF of Clade II, which led to a decrease in virulence. In the case of Clade I, it is based on the inhibition of T-cell activation, which suppresses the expression of pro-inflammatory cytokines [1, 16]. The current world outbreak (2022–2023) was identified as Clade IIb, a subtype of the West African clade with low severity, presumably exported out of Africa and amplified by travelers who had sexual contact with men [2]. Currently, most infected people belong to the B.1 lineage of Clade IIb, and some belong to the A.2 lineage. Lineage B.1 has accumulated over 50 nucleotide changes compared to a Clade IIb MPXV isolated in 2018 [17]. Although the total fatality rate of cases counted after the outbreak was significantly lower at 0.16% (deaths/confirmed cases:140/87,479), these many nucleotide changes are likely to have influenced the activity alteration of APOBEC3 cytosine deaminase and the substantial increase in human-to-human transmission [18].
Diagnosis
The analytical methods developed for the detection of MPXV are typically conducted in biosafety level 3 facilities and are similar to the methods for other viruses. The most efficient method for sample preservation and fast analysis involves real-time polymerase chain reaction (PCR) using viral DNA isolated from lesion tissue. Since MPXV is rarely detected except during the short period of initial infection, the PCR analysis using serum samples is not recommended by the WHO [19]. Additionally, methods for detecting viral proteins and antibodies in serum are prioritized and widely utilized. Electron microscopy analysis of viruses isolated from clinical specimens and immunohistochemistry of tissue samples are also available, but they have limitations, as they require skilled techniques and specialized analytical equipment.
The diagnostic targets of MPXV are diverse. Among MPXV genes, the D6R gene of MPXV is a conserved region (> 100 nucleotides in length) that spans the entire
As an immunological method in clinical area, enzyme-linked immunosorbent assay (ELISA) which has been developed to detect patient antibodies (IgG, IgM) against Orthopoxvirus is still used to MPXV as well in the serum at five days or more than eight days after the onset of rash [27, 28]. However, the detection specificity of these antibodies is insufficient to distinguish because of the highly conserved antigen epitopes between MPXV and other poxviruses [1]. In addition, ELISA for detecting IgG and IgM has the disadvantage of being unable to distinguish between people infected with other poxviruses in the past, people who have been vaccinated, and people infected with Mpox [1, 27, 29]. Consequently, the use of this method is limited to epidemiologic studies [30].
Antiviral Therapies
Currently, no specific antiviral therapy has been approved for treating human MPXV infections. During the ongoing Mpox outbreak, supportive care and pain control are recommended as the initial treatment options for most patients. Owing to their broad-spectrum antiviral activity, several therapies (cidofovir, brincidofovir, tecovirimat, and vaccinia immunoglobulin) for smallpox infection are considered for the treatment of severe cases of Mpox and high-risk patients such as immunocompromised people, infants, pregnant or breastfeeding women, and people with skin integrity issues (Treatment Information for Healthcare Professionals from CDC [31]).
A viral replication step has been the most important antiviral target as nucleotide analogs can interfere synthesis of viral genomes. Cidofovir, an FDA-approved drug used to treat cytomegalovirus infections, inhibits DNA synthesis and interferes with replication of various DNA viruses. Owing to its broad-spectrum activity, cidofovir is considered a potential therapeutic option against other DNA viruses, including orthopoxviruses. Cidofovir remains an investigational treatment option for smallpox [32] and Mpox. Brincidofovir is a cidofovir prodrug that contains a lipid conjugate for better membrane permeability in infected cells [33]. Brincidofovir showed higher protective efficacy than cidofovir against various DNA viruses, such as orthopoxviruses [34], herpesviruses [35], and adenoviruses [36]. Recently, FDA approved brincidofovir for the treatment of smallpox. Both cidofovir and brincidfovir are nucleoside analogs that are incorporated into newly synthesized DNA strands, terminating DNA chain elongation [37].
Viral envelope is another important antiviral drug target as it plays a critical role not only in maintaining virion and protecting from diverse environment but also in viral entry. Tecovirimat, another FDA-approved drug used to treat smallpox, targets a specific orthopoxvirus membrane protein p37 (F13L of VACV), which is responsible for forming the envelope of immature intracellular virus particles [38], eventually interfering with cell-to-cell and systemic virus transmission [37, 39]. Since p37 is highly conserved in orthopoxviruses, the p37-targeting drug tecovirimat is considered a potential therapeutic option for ongoing Mpox outbreaks and is currently undergoing various clinical trials.
Each stage of the viral life cycle may be considered a potential target for the development of new antiviral drugs. Many investigational anti-orthopoxviral drugs that target key components at different stages, from viral entry to progeny release, have been identified (Table 1).
-
Table 1 . Candidate anti-orthopoxvirus agents with defined targets..
Stage of life cycle Candidate agent Mechanism of action Stage of development Ref Entry Vaccinia immune globulin Neutralizing viral particles Approved for vaccinia infection [40, 41] Early transcription Adenosine N1-oxide Nigericin
Aurintricarboxylic acidInhibiting viral early gene transcription Preclinical research [42]
[43]
[44]DNA synthesis Cidofovir Nucleoside analogs Available through IND* protocol for smallpox [37] Brincidofovir Approved for smallpox / Clinical trial phase 3 for dsDNA viruses including MPOX [45] N-Methanocarbathymidine
Aphidicolin
Cytosine arabinoside
Phosphonoacetic acid
4'-thioIDU
5-substituted deoxyuridine analogsPreclinical research [46]
[47]
[48]
[48]
[49]
[50]Small peptide aptamers Binding to virus replication complex Preclinical research [51, 52] Hydroxyurea Ribonucleotide reductase inhibitor Preclinical research [53] Late transcription IBT
Methisazone
Ethacrynic acid
Alpha-lipoic acidInhibiting viral late gene transcription Preclinical research [54, 55]
[57]
[58]
[58]Assembly Rifampicin Interfering formation of viral membrane Preclinical research [59, 60] Mitoxantrone DNA ligase inhibitor Preclinical research [61, 62] Ofloxacin
NovobiocinTopoisomerase inhibitor Preclinical research [63]
[64, 65]Maturation TTP-6171 Cysteine proteinase inhibitor Preclinical research [66] Secondary envelopment Tecovirimat VP37 inhibitor Approved for smallpox / Clinical trial phase 3 for MPOX [68] IMCBH Preventing intracellular virion wrapping Preclinical research [70, 71] Egress Imatinib Inhibiting cellular
Abl-family tyrosine kinasesPreclinical research [72-74] Terameprocol Inhibiting actin tail formation Preclinical research [75] Nitroxoline Inhibiting the signal cascade that modulates virus replication Preclinical research [76-78] *Investigational new drug (IND).
I. Entry and uncoating
• Vaccinia immune globulin [40, 41]: Neutralizing activity of antibodies blocks the interaction between virions and host receptors, inhibiting viral entry.
II. Early transcription
• Adenosine N1-oxide [42]: Incorporation of adenosine N1-oxide into early viral transcripts interferes with the translation of viral mRNA, not cellular mRNA, blocking early viral gene expression.
• Nigericin [43]: Although its precise mechanism of action remains unknown, nigericin inhibits viral replication at the early transcription stage. As a carboxylic ionophore, nigericin can be readily incorporated into biological membranes to stimulate the exchange of monovalent cations with protons. These environmental changes may affect the viral mRNA synthesis.
• Aurintricarboxylic acid [44]: Aurintricarboxylic acid inhibits the transcription of early viral genes by targeting both the cellular (blocking the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling cascade, which is essential for VACV replication) and viral factors (inhibiting the phosphatase activity of the viral enzyme H1L, which is required to initiate viral transcription).
III. DNA synthesis
• Nucleoside analogs: Nucleoside analogs mimic natural nucleosides to compete with natural nucleosides during DNA synthesis and are incorporated into newly synthesized DNA strands. Once incorporated, nucleoside analogs terminate the DNA chain elongation. Numerous nucleoside analogs have been identified as antiviral therapeutics, and several compounds have been confirmed to induce broad-spectrum anti-orthopoxviral activities (listed below).
◆ Cidofovir & brincidofovir [37, 45]
◆ N-methanocarbathymidine [46]
◆ Aphidicolin [47]
◆ Cytosine arabinoside & phosphonoacetic acid [48]
◆ 1-(2-deoxy-4-thio-beta-d-ribofuranosyl)-5-iodouracil (4'-thioIDU) [49]
◆ 5-substituted deoxyuridine analogs [50]: Viral thymidine kinase phosphorylates these modified molecules, enhancing their antiviral activities.
• Small peptide aptamers [51, 52]: Peptide aptamers targeting A20, a major component of the viral replication complex, impair viral DNA synthesis.
• Hydroxyurea [53] is a ribonucleotide reductase inhibitor that limits deoxyribonucleotide production, thereby inhibiting viral DNA synthesis.
IV. Late transcription
• Isatin-beta-thiosemicarbazone (IBT) [54, 55]: The precise mechanism of action of IBT is not completely understood. However, IBT terminates late viral gene transcripts by degrading late viral mRNAs, followed by the cessation of late viral protein production. Moreover, IBT indirectly regulates viral transcription elongation factors [56].
• Methisazone (N-methyl-isatin-/i-thiosemicarbazone, N-MeIBT,MelBT) [57]: Methisazone inhibits mRNA and protein synthesis and has antiviral activity. Methisazone has been reported to be effective in smallpox prophylaxis.
• Ethacrynic and alpha-lipoic acid [58]: Both ethacrynic and alpha-lipoic acids inhibit vaccinia late gene expression, but not viral entry, early gene expression, and viral DNA synthesis.
V. Assembly
• Rifampicin [59, 60]: Rifampicin interferes with the formation of viral membranes during the early assembly step, resulting in the inhibition of viral morphogenesis.
• Mitoxantrone [61, 62]: As a DNA ligase inhibitor, mitoxantrone blocks virion assembly, but its protective efficacy is limited in vivo.
• Ofloxacin [63]: Ofloxacin inhibits topoisomerase, which is essential for virion assembly.
• Novobiocin [64, 65]: Novobiocin is also a topoisomerase inhibitor, interfering with virion assembly.
VI. Maturation
• TTP-6171 [66]: TTP-6171 inhibits I7L core proteinase, a cysteine proteinase that processes viral structural and membrane proteins, and blocks the maturation of intracellular virions.
• Recently, a computer modeling strategy identified several potential protease inhibitors with better binding affinity than that of TTP-6171 [67]: (1) Gallicynoic Acid F (NPA002071), (2) H2-Erythro-Neopterin (NPA000530), (3) Nigcollin C (NPA029767), (4) NPA24545, and (5) Vaccinol M (NPA030378)
VII. Secondary envelopment
• Tecovirimat [68]: A VP37-targeting agent tecovirimat inhibits the formation of enveloped viral particles. Formation of this envelope is important for viral egress and the next round of infection in other cells. Accordingly, targeting VP37 with a tecovirimat prevents the viral spread to other cells in the body. Its protective efficacy against the circulating Mpox strain has been confirmed in vitro [69] and clinical trials are in progress.
• N1-isonicotinoyl-N2-3-methyl-4-chloro-benzoylhydrazine (IMCBH) [70, 71]: IMCBH is another agent that affects secondary envelope formation by preventing the wrapping of intracellular virions by Golgi membranes.
VIII. Release of progeny virus particles
• Imatinib [72-74]: As a host-targeting agent, imatinib inhibits cellular Abl family tyrosine kinases, which are required for the release of infectious progeny viral particles.
• Terameprocol [75]: Terameprocol interferes the formation of actin tails which are crucial for spreading viruses to neighboring cells.
• Host targeting drug, nitroxoline [76]: Although its mechanism of action is not known, nitroxoline inhibits the phosphoinositide 3‐kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling pathway and stimulates the fibrosarcoma/mitogen‐activated protein kinase/extracellular signal‐regulated kinase (RAF/MEK/ERK) signaling pathway. Both signaling cascades are involved in orthopoxvirus replication [77, 78].
Vaccines
The currently available smallpox vaccine Dryvax, a live VACV that protects humans from both smallpox and Mpox, is a cloned vaccine strain [79]. However, large doses of these vaccines can cause serious side effects in high-risk individuals with acquired or congenital immune system defects [80]. Interestingly, the main mode of protection of an unattenuated smallpox vaccine against Mpox is mediated by neutralizing antibodies (Abs) [81]. Unattenuated Dryvax, modified vaccinia Ankara (MVA), and NYVAC do not protect against lethal Mpox in immunodeficient animals infected with the simian immunodeficiency virus (SIV) [82]. This was confirmed to be due to defective maturation of high-affinity protective Abs under CD4+ T cell depletion. LC16m8 [83], a VACV-mediated vaccine attenuated by passaging in mammalian primary cell, and VACΔ6 [84], a VACV-mediated vaccine intentionally modified six genes, also remain in phase 2 and 1 clinical trials respectively as a vaccine against MPXV yet (All clinical status was referenced in the ‘WHO Mpox Vaccine Tracker’ updated on April 24, 2023 [85]).
Orthopoxviruses, a family of double-stranded DNA viruses, comprise a variety of viruses, including human MPXV, variola virus, VACV, and cowpox virus [86]. Orthopoxvirus infection and immunization can confer immunity to other viruses, and as a result, two vaccines have been approved by the FDA to prevent MPXV [87]. ACAM2000 is a second-generation live VACV vaccine, and Bavarian Nordic (JYNNEOS, also known as MVA-BN, Imvamune, or Imvanex) was developed as a modified vaccinia Ankara (MVA)-based attenuated third-generation smallpox vaccine [88]. Since approval, the indication has been expanded based on the data from the MPXV challenge study conducted in non-humans (FDA's authorized “JYNNEOS” vaccine for counteracting Mpox global public health emergency). These two VACV-based smallpox vaccines cross-protect against MPXV [6]. Unfortunately, smallpox vaccines do not completely protect against circulating MPXV [89]. ACAM2000 is a highly regenerated VACV vaccine with serious side effects [90] (Table 2). In contrast, JYNNEOS, which is based on a live attenuated orthopoxvirus, is expected to have the lowest incidence of severe adverse events as a replication-deficient vaccine [90].
-
Table 2 . Approved vaccines to prevent MPXV infection..
Trade name Manufacturer Molecule type Target disease Inoculation site Dosing interval and number Injection volume Side effects JYNNEOS/ Imvanex Bavarian
Nordic ASLive
Attenuated
VaccineMonkeypox; smallpox; vaccinia virus (EU only) Subcutaneous
Intradermal2 doses administered 28 days (4 weeks) apart 0.5 ml
0.1 mlPain at the injection site, redness, itching, fever, headache, tiredness, nausea, chills, and muscle aches ACAM2000 Gaithersburg, Inc. Live virus
VaccineSmallpox Percutaneous Single dose using a bifurcated needle 0.0025 ml droplet of reconstituted vaccine (100 doses) Itching, sore arm, fever, headache, body ache, mild rash, fatigue, myocarditis, and pericarditis
Development of Novel Mpox Vaccines
Currently, mRNA- and DNA-based vaccines are being developed. Among them, one of the mRNA vaccine is the combination of mRNA-A-LNP and mRNA-B-LNP which are designed to contain two IMV-specific proteins (A29L and M1R) and two infected EEV-specific proteins (A35R and B6R) of MPXV, respectively [91]. Combinations of IMV- and EEV-specific immunogens have been studied to provide more protection than immunogens alone [92]. In a recent study, the immunogenicity of a multivalent mRNA vaccine candidate, MPXVac-097 (A29L, A35R, M1R, E8L, and B6R linked to a 2a peptide), was characterized in a mouse model [93]. MPXVac-097 elicits MPXV-specific T-cell responses and neutralizes VACV infections. As other effective mRNA vaccine candidates against MPXV, Rmix4 and Rmix6 encode each four (M1R, A29L, B6R, A35R) and six (M1R, H3L, A29L, E8L, B6R, and A35R) of MPXV antigens [94]. Freyn
Alternatively, several VACV genes and gene combinations have been tested for their immunogenicity and protective efficacy in mice using a gene gun delivery DNA vaccine. [97]. The four-gene combination DNA vaccine showed immunogenicity in non-human primates and protected 100% of the VACV-infected mice. DNA vaccines (expressing A27L, A33R, L1R, and B5R), named 4pox, have been reported to protect against fatal Mpox, rabbit pox, and VACV infections [98, 99]. Interestingly, vaccination of plasmid DNAs encoding these four antigens (L1R, A27L, A33R, and B5R) with boosting by the equivalent recombinant proteins showed much improved vaccine efficacy against MPXV [100]. In addition, Hirao
Franceschi
Conventional recombinant protein-based vaccines are currently being developed. Because live poxviral vectors express many cytokines and chemokines, it is difficult to analyze the effects of immunomodulatory approaches [103]. On the other hands, vaccination of the recombinant subunit platform comprising A33R, B5R, L1R, and A27L of VACV with Alum adjuvant and CpG was reported successful IgG generation and full protection against VACV, even in a single-targeting vaccination trials [104, 105]. This study presents a simple subunit-based vaccine with excellent potential to increase immunogenicity and protective efficacy in high-risk populations.
Synthetic vaccines have recently been proposed as new tools for controlling infectious diseases. The novel immunogens containing a mixture of synthetic peptides, which were site-specifically designed for epitopes exposed on the multiple proteins of pathogen, has been shown to be an efficient approach as vaccine development [106]. Importantly, these synthetic vaccines promote the production of antibodies that neutralize the viral proteins when administered in monkeys and were able to delay or suppress the disease development in immunized human volunteers [107]. This strategy can be reliably used for diseases caused by viruses such as MPXV by including immunogenic epitopes and appropriate immune stimulants [108]. This allows development at a relatively low cost, particularly owing to low biosafety requirements, and presents remarkable immune diversity [109].
-
Figure 3. The characteristics of vaccine candidates against Mpox.
Summary
Currently available therapeutics and vaccines for smallpox are cross-reactive to MPXV [6, 31, 32]. However, there are limitations to the therapeutic efficacy of different treatment and vaccine products against Mpox; thus, the development of specific therapeutics and vaccines against Mpox is still required [89]. One of them is that VACV exist in two forms (IMV and EEV) and infect cells by different mechanisms [11]. Thus, it is highly recommended to develop antiviral drugs targeting the replication steps of the viral life cycle, which can inhibit viral propagation regardless of viral entry. For example, tecovirimat does not affect the intracellular form of the virus and can only inhibit the formation of enveloped virions because it targets the p37 membrane protein of VACV, which only exists in EEV [68, 110]. In contrast, cidofovir and brincidofovir block orthopoxvirus DNA polymerase-mediated DNA synthesis, inhibiting viral replication [37, 45].
As for the vaccine development, multivalent vaccines created by the addition of MPXV immunogens to previously developed vaccine components are key strategies for the development of Mpox vaccines. Since two forms of VACV (i.e., IMV and EEV) can enter cells by different mechanisms, the composition of immunogens may affect their neutralizing activity. Indeed, despite the high immunogenicity, sera from animals immunized with the EEV proteins did not show neutralizing activity against IMV. More importantly, protective efficacy was better in animals immunized with combinations of IMV and EEV proteins than in animals immunized with individual IMV or EEV proteins [111]. Hence, this should be considered in the development of multivalent Mpox vaccines.
Another key strategy for improving vaccines is to strengthen previously developed vaccines to cover more diverse strains of orthopoxviruses, including Mpox. This is possible by boosting cellular immune responses. Since Mpox has highly similar amino acid sequences to vaccinia viruses, T cell-mediated cellular responses induced by VACV vaccines are cross-reactive to Mpox [112]. Optimization of adjuvants or the addition of peptides conserved between the VACV and Mpox can increase T cell-mediated cellular responses, allowing vaccines to target a broad range of viruses.
Together with the development of rapid diagnostics specific for Mpox, improvements in the effectiveness of therapeutics and vaccines against Mpox will prevent future Mpox outbreaks.
Acknowledgments
This work was supported in part by the Korea Research Institute of Chemical Technology (KRICT) (Project No. KK2333-20) and the National Research Foundation of Korea (grant number NRF-2021M3E5E3080540).
Conflict of Interest
The authors have no financial conflicts of interest to declare.
Fig 1.
Fig 2.
Fig 3.
-
Table 1 . Candidate anti-orthopoxvirus agents with defined targets..
Stage of life cycle Candidate agent Mechanism of action Stage of development Ref Entry Vaccinia immune globulin Neutralizing viral particles Approved for vaccinia infection [40, 41] Early transcription Adenosine N1-oxide Nigericin
Aurintricarboxylic acidInhibiting viral early gene transcription Preclinical research [42]
[43]
[44]DNA synthesis Cidofovir Nucleoside analogs Available through IND* protocol for smallpox [37] Brincidofovir Approved for smallpox / Clinical trial phase 3 for dsDNA viruses including MPOX [45] N-Methanocarbathymidine
Aphidicolin
Cytosine arabinoside
Phosphonoacetic acid
4'-thioIDU
5-substituted deoxyuridine analogsPreclinical research [46]
[47]
[48]
[48]
[49]
[50]Small peptide aptamers Binding to virus replication complex Preclinical research [51, 52] Hydroxyurea Ribonucleotide reductase inhibitor Preclinical research [53] Late transcription IBT
Methisazone
Ethacrynic acid
Alpha-lipoic acidInhibiting viral late gene transcription Preclinical research [54, 55]
[57]
[58]
[58]Assembly Rifampicin Interfering formation of viral membrane Preclinical research [59, 60] Mitoxantrone DNA ligase inhibitor Preclinical research [61, 62] Ofloxacin
NovobiocinTopoisomerase inhibitor Preclinical research [63]
[64, 65]Maturation TTP-6171 Cysteine proteinase inhibitor Preclinical research [66] Secondary envelopment Tecovirimat VP37 inhibitor Approved for smallpox / Clinical trial phase 3 for MPOX [68] IMCBH Preventing intracellular virion wrapping Preclinical research [70, 71] Egress Imatinib Inhibiting cellular
Abl-family tyrosine kinasesPreclinical research [72-74] Terameprocol Inhibiting actin tail formation Preclinical research [75] Nitroxoline Inhibiting the signal cascade that modulates virus replication Preclinical research [76-78] *Investigational new drug (IND).
-
Table 2 . Approved vaccines to prevent MPXV infection..
Trade name Manufacturer Molecule type Target disease Inoculation site Dosing interval and number Injection volume Side effects JYNNEOS/ Imvanex Bavarian
Nordic ASLive
Attenuated
VaccineMonkeypox; smallpox; vaccinia virus (EU only) Subcutaneous
Intradermal2 doses administered 28 days (4 weeks) apart 0.5 ml
0.1 mlPain at the injection site, redness, itching, fever, headache, tiredness, nausea, chills, and muscle aches ACAM2000 Gaithersburg, Inc. Live virus
VaccineSmallpox Percutaneous Single dose using a bifurcated needle 0.0025 ml droplet of reconstituted vaccine (100 doses) Itching, sore arm, fever, headache, body ache, mild rash, fatigue, myocarditis, and pericarditis
References
- McCollum AM, Damon IK. 2013. Human Monkeypox.
Clin. Infect. Dis. 58 : 260-267. - WHO. 2023. 2022-23 Mpox outbreak: Global Trends. Available from https://worldhealthorg.shinyapps.io/mpx_global/. Accessed on May 16, 2023.
- Chadha J, Khullar L, Gulati P, Chhibber S, Harjai K. 2022. Insights into the monkeypox virus: Making of another pandemic within the pandemic?
Environ. Microbiol. 24 : 4547-4560. - 2023. Mpox (Monkeypox): Causes, Symptoms, Treatment & Prevention. Available from https://my.clevelandclinic.org/health/diseases/22371-monkeypox#symptoms-and-causes. Accessed on April 25, 2023.
- 2022. Investigation into monkeypox outbreak in England: technical briefing 1. Available from https://www.gov.uk/government/publications/monkeypox-outbreak-technical-briefings/investigation-into-monkeypox-outbreak-in-england-technical-briefing-1#sources-and-acknowledgments. Accessed on Sep. 23, 2023.
- Ahmed SF, Sohail MS, Quadeer AA, McKay MR. 2022. Vaccinia-virus-based vaccines are expected to elicit highly cross-reactive immunity to the 2022 monkeypox virus.
Viruses 14 : 1960. - Louten J, Reynolds N. 2016. Essential human virology, pp. 273-290. Ed. Academic Press is an imprint of Elsevier, London, UK.
- Shi D, He P, Song Y, Cheng S, Linhardt RJ, Dordick JS,
et al . 2022. Kinetic and structural aspects of glycosaminoglycan - Monkeypox virus protein A29 interactions using surface plasmon resonance.Molecules 27 : 5898. - Moss B. 2016. Membrane fusion during poxvirus entry.
Semin. Cell. Dev. Biol. 60 : 89-96. - Roberts KL, Breiman A, Carter GC, Ewles HA, Hollinshead M, Law M,
et al . 2009. Acidic residues in the membrane-proximal stalk region of vaccinia virus protein B5 are required for glycosaminoglycan-mediated disruption of the extracellular enveloped virus outer membrane.J. Gen. Virol. 90 : 1582-1591. - Gray RDM, Albrecht D, Beerli C, Huttunen M, Cohen GH, White IJ,
et al . 2019. Nanoscale polarization of the entry fusion complex of vaccinia virus drives efficient fusion.Nat. Microbiol. 4 : 1636-1644. - Moss B. 2013. Poxvirus DNA replication.
Cold Spring Harb. Perspect. Biol. 5 : a010199. - Katsafanas GC, Moss B. 2007. Colocalization of transcription and translation within cytoplasmic poxvirus factories coordinates viral expression and subjugates host functions.
Cell Host Microbe. 2 : 221-228. - Li H, Huang QZ, Zhang H, Liu ZX, Chen XH, Ye LL,
et al . 2023. The land-scape of immune response to monkeypox virus.EBioMedicine 87 : 104424. - Liang C, Qian J, Liu L. 2022. Biological characteristics, biosafety prevention and control strategies for the 2022 multi-country outbreak of monkeypox.
Biosaf. Health 04 : 376-385. - Reynolds MG, Damon IK. 2012. Outbreaks of human monkeypox after cessation of smallpox vaccination.
Trends Microbiol. 20 : 80-87. - Isidro J, Borges V, Pinto M, Sobral D, Santos JD, Nunes A,
et al . 2022. Phylogenomic characterization and signs of microevolution in the 2022 multi-country outbreak of monkeypox virus.Nat. Med. 28 : 1569-1572. - Alcami A. 2023. Pathogenesis of the circulating mpox virus and its adaptation to humans.
Proc. Natl. Acad. Sci. USA 120 : e2301662120. - WHO. 2022. Laboratory testing for the monkeypox virus: Interim guidance. Available from https://www.who.int/publications/i/item/WHO-MPX-laboratory-2022.1. Accessed on 05.23, 2022.
- Luciani L, Inchauste L, Ferraris O, Charrel R, Nougairede A, Piorkowski G,
et al . 2021. A novel and sensitive real-time PCR system for universal detection of poxviruses.Sci. Rep. 11 : 1798. - Orba Y, Sasaki M, Yamaguchi H, Ishii A, Thomas Y, Ogawa H,
et al . 2015. Orthopoxvirus infection among wildlife in Zambia.J. Gen. Virol. 96 : 390-394. - Li Y, Olson VA, Laue T, Laker MT, Damon IK. 2006. Detection of monkeypox virus with real-time PCR assays.
J. Clin. Virol. 36 : 194-203. - Shchelkunov SN, Shcherbakov DN, Maksyutov RA, Gavrilova EV. 2011. Species-specific identification of variola, monkeypox, cowpox, and vaccinia viruses by multiplex real-time PCR assay.
J. Virol. Methods 175 : 163-169. - Kulesh DA, Loveless BM, Norwood D, Garrison J, Whitehouse CA, Hartmann C,
et al . 2004. Monkeypox virus detection in rodents using real-time 3'-minor groove binder TaqMan assays on the Roche LightCycler.Lab. Invest. 84 : 1200-1208. - CDC. 2023. Mpox Generic PCR Protocol. Available from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on March 7, 2023.
- Li Y, Zhao H, Wilkins K, Hughes C, Damon IK. 2010. Real-time PCR assays for the specific detection of monkeypox virus West African and Congo Basin strain DNA.
J. Virol. Methods 169 : 223-227. - WHO. 2022. Surveillance, case investigation and contact tracing for mpox (monkeypox): interim guidance. Available from https://www.who.int/publications/i/item/WHO-MPX-Surveillance-2022.4. Accessed on 12.22, 2022.
- Karem KL, Reynolds M, Braden Z, Lou G, Bernard N, Patton J,
et al . 2005. characterization of acute-phase humoral immunity to monkeypox: use of immunoglobulin M enzyme-linked immunosorbent assay for detection of monkeypox infection during the 2003 North American outbreak.Clin. Diagn. Lab. Immunol. 12 : 867-872. - Karem KL, Reynolds M, Braden Z, Lou G, Bernard N, Patton J,
et al . 2005. Characterization of acute-phase humoral immunity to Monkeypox: Use of immunoglobulin M enzyme-linked immunosorbent assay for detection of Monkeypox infection during the 2003 North American outbreak.Clin. Diagn. Lab. Immunol. 12 : 867-872. - Alakunle E, Moens U, Nchinda G, Okeke MI. 2020. Monkeypox virus in Nigeria: Infection biology, epidemiology, and evolution.
Viruses 12 : 1257. - CDC. 2023. Treatment Information for Healthcare Professionals. Available from https://www.cdc.gov/poxvirus/mpox/clinicians/treatment.html. Accessed vailable from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on April 21, 2023.
- FDA. 2023. Smallpox Preparedness and Response Updates from FDA. Available from https://www.fda.gov/emergencypreparedness-and-response/mcm-issues/smallpox-preparedness-and-response-updates-fda. Accessed vailable from https://www.cdc.gov/poxvirus/mpox/lab-personnel/index.html. Accessed on May 3, 2023.
- Hostetler KY. 2009. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art.
Antiviral Res. 82 : A84-98. - Kern ER, Hartline C, Harden E, Keith K, Rodriguez N, Beadle JR,
et al . 2002. Enhanced inhibition of orthopoxvirus replication in vitro by alkoxyalkyl esters of cidofovir and cyclic cidofovir.Antimicrob. Agents Chemother. 46 : 991-995. - Beadle JR, Hartline C, Aldern KA, Rodriguez N, Harden E, Kern ER,
et al . 2002. Alkoxyalkyl esters of cidofovir and cyclic cidofovir exhibit multiple-log enhancement of antiviral activity against cytomegalovirus and herpesvirus replication in vitro.Antimicrob. Agents Chemother. 46 : 2381-2386. - Hartline CB, Gustin KM, Wan WB, Ciesla SL, Beadle JR, Hostetler KY,
et al . 2005. Ether lipid-ester prodrugs of acyclic nucleoside phosphonates: activity against adenovirus replication in vitro.J. Infect. Dis. 191 : 396-399. - Siegrist EA, Sassine J. 2023. Antivirals with activity against Mpox: A clinically oriented review.
Clin. Infect. Dis. 76 : 155-164. - Yang G, Pevear DC, Davies MH, Collett MS, Bailey T, Rippen S,
et al . 2005. An orally bioavailable antipoxvirus compound (ST-246) inhibits extracellular virus formation and protects mice from lethal orthopoxvirus challenge.J. Virol. 79 : 13139-13149. - Grosenbach DW, Jordan R, Hruby DE. 2011. Development of the small-molecule antiviral ST-246 as a smallpox therapeutic.
Future Virol. 6 : 653-671. - Bell E, Shamim M, Whitbeck JC, Sfyroera G, Lambris JD, Isaacs SN. 2004. Antibodies against the extracellular enveloped virus B5R protein are mainly responsible for the EEV neutralizing capacity of vaccinia immune globulin.
Virology 325 : 425-431. - Thet AK, Kelly PJ, Kasule SN, Shah AK, Chawala A, Latif A,
et al . 2023. The use of vaccinia immune globulin in the treatment of severe Mpox. virus infection in human immunodeficiency virus/AIDS.Clin. Infect. Dis. 76 : 1671-1673. - Kane EM, Shuman S. 1995. Adenosine N1-oxide inhibits vaccinia virus replication by blocking translation of viral early mRNAs.
J. Virol. 69 : 6352-6358. - Myskiw C, Piper J, Huzarewich R, Booth TF, Cao J, He R. 2010. Nigericin is a potent inhibitor of the early stage of vaccinia virus replication.
Antiviral Res. 88 : 304-310. - Myskiw C, Deschambault Y, Jefferies K, He R, Cao J. 2007. Aurintricarboxylic acid inhibits the early stage of vaccinia virus replication by targeting both cellular and viral factors.
J. Virol. 81 : 3027-3032. - Rizk JG, Lippi G, Henry BM, Forthal DN, Rizk Y. 2022. Prevention and treatment of Monkeypox.
Drugs 82 : 957-963. - Prichard MN, Keith KA, Quenelle DC, Kern ER. 2006. Activity and mechanism of action of
N -methanocarbathymidine against herpesvirus and orthopoxvirus infections.Antimicrob. Agents Chemother. 50 : 1336-1341. - Pedrali-Noy G, Spadari S. 1980. Mechanism of inhibition of herpes simplex virus and vaccinia virus DNA polymerases by aphidicolin, a highly specific inhibitor of DNA replication in eucaryotes.
J. Virol. 36 : 457-464. - Taddie JA, Traktman P. 1993. Genetic characterization of the vaccinia virus DNA polymerase: cytosine arabinoside resistance requires a variable lesion conferring phosphonoacetate resistance in conjunction with an invariant mutation localized to the 3'-5' exonuclease domain.
J. Virol. 67 : 4323-4336. - Kern ER, Prichard MN, Quenelle DC, Keith KA, Tiwari KN, Maddry JA,
et al . 2009. Activities of certain 5-substituted 4'-thiopyrimidine nucleosides against orthopoxvirus infections.Antimicrob. Agents Chemother. 53 : 572-579. - Smee DF, Humphreys DE, Hurst BL, Barnard DL. 2008. Antiviral activities and phosphorylation of 5-halo-2'-deoxyuridines and Nmethanocarbathymidine in cells infected with vaccinia virus.
Antivir. Chem. Chemother. 19 : 15-24. - Saccucci L, Crance JM, Colas P, Bickle M, Garin D, Iseni F. 2009. Inhibition of vaccinia virus replication by peptide aptamers.
Antiviral Res. 82 : 134-140. - Flusin O, Saccucci L, Contesto-Richefeu C, Hamdi A, Bardou C, Poyot T,
et al . 2012. A small molecule screen in yeast identifies inhibitors targeting protein-protein interactions within the vaccinia virus replication complex.Antiviral Res. 96 : 187-195. - Slabaugh MB, Mathews CK. 1986. Hydroxyurea-resistant vaccinia virus: overproduction of ribonucleotide reductase.
J. Virol. 60 : 506-514. - Condit RC, Easterly R, Pacha RF, Fathi Z, Meis RJ. 1991. A vaccinia virus isatin-beta-thiosemicarbazone resistance mutation maps in the viral gene encoding the 132-kDa subunit of RNA polymerase.
Virology 185 : 857-861. - Meis RJ, Condit RC. 1991. Genetic and molecular biological characterization of a vaccinia virus gene which renders the virus dependent on isatin-beta-thiosemicarbazone (IBT).
Virology 182 : 442-454. - Prins C, Cresawn SG, Condit RC. 2004. An isatin-beta-thiosemicarbazone-resistant vaccinia virus containing a mutation in the second largest subunit of the viral RNA polymerase is defective in transcription elongation.
J. Biol. Chem. 279 : 44858-44871. - Weiss MM, Weiss PD, Mathisen G, Guze P. 2004. Rethinking smallpox.
Clin. Infect. Dis. 39 : 1668-1673. - Spisakova M, Cizek Z, Melkova Z. 2009. Ethacrynic and alpha-lipoic acids inhibit vaccinia virus late gene expression.
Antiviral Res. 81 : 156-165. - Charity JC, Katz E, Moss B. 2007. Amino acid substitutions at multiple sites within the vaccinia virus D13 scaffold protein confer resistance to rifampicin.
Virology 359 : 227-232. - Baldick CJ Jr., Moss B. 1987. Resistance of vaccinia virus to rifampicin conferred by a single nucleotide substitution near the predicted NH2 terminus of a gene encoding an Mr 62,000 polypeptide.
Virology 156 : 138-145. - Deng L, Dai P, Ciro A, Smee DF, Djaballah H, Shuman S. 2007. Identification of novel antipoxviral agents: mitoxantrone inhibits vaccinia virus replication by blocking virion assembly.
J. Virol. 81 : 13392-13402. - Altmann SE, Smith AL, Dyall J, Johnson RF, Dodd LE, Jahrling PB,
et al . 2012. Inhibition of cowpox virus and monkeypox virus infection by mitoxantrone.Antiviral Res. 93 : 305-308. - Ikeda S, Yazawa M, Nishimura C. 1987. Antiviral activity and inhibition of topoisomerase by ofloxacin, a new quinolone derivative.
Antiviral Res. 8 : 103-113. - Sekiguchi J, Stivers JT, Mildvan AS, Shuman S. 1996. Mechanism of inhibition of vaccinia DNA topoisomerase by novobiocin and coumermycin.
J. Biol. Chem. 271 : 2313-2322. - Sekiguchi J, Shuman S. 1997. Novobiocin inhibits vaccinia virus replication by blocking virus assembly.
Virology 235 : 129-137. - Byrd CM, Bolken TC, Mjalli AM, Arimilli MN, Andrews RC, Rothlein R,
et al . 2004. New class of orthopoxvirus antiviral drugs that block viral maturation.J. Virol. 78 : 12147-12156. - Dubey A, Alawi MM, Alandijany TA, Alsaady IM, Altwaim SA, Sahoo AK,
et al . 2023. Exploration of microbially derived natural compounds against Monkeypox virus as viral core cysteine proteinase inhibitors.Viruses 15 : 251. - Prichard MN, Kern ER. 2012. Orthopoxvirus targets for the development of new antiviral agents.
Antiviral Res. 94 : 111-125. - Frenois-Veyrat G, Gallardo F, Gorge O, Marcheteau E, Ferraris O, Baidaliuk A,
et al . 2022. Tecovirimat is effective against human monkeypox virus in vitro at nanomolar concentrations.Nat. Microbiol. 7 : 1951-1955. - Payne LG, Kristenson K. 1979. Mechanism of vaccinia virus release and its specific inhibition by N1-isonicotinoyl-N2-3-methyl-4-chlorobenzoylhydrazine.
J. Virol. 32 : 614-622. - Schmutz C, Payne LG, Gubser J, Wittek R. 1991. A mutation in the gene encoding the vaccinia virus 37,000-M(r) protein confers resistance to an inhibitor of virus envelopment and release.
J. Virol. 65 : 3435-3442. - Reeves PM, Smith SK, Olson VA, Thorne SH, Bornmann W, Damon IK,
et al . 2011. Variola and monkeypox viruses utilize conserved mechanisms of virion motility and release that depend on abl and SRC family tyrosine kinases.J. Virol. 85 : 21-31. - Newsome TP, Weisswange I, Frischknecht F, Way M. 2006. Abl collaborates with Src family kinases to stimulate actin-based motility of vaccinia virus.
Cell. Microbiol. 8 : 233-241. - Reeves PM, Bommarius B, Lebeis S, McNulty S, Christensen J, Swimm A,
et al . 2005. Disabling poxvirus pathogenesis by inhibition of Abl-family tyrosine kinases.Nat. Med. 11 : 731-739. - Pollara JJ, Laster SM, Petty IT. 2010. Inhibition of poxvirus growth by Terameprocol, a methylated derivative of nordihydroguaiaretic acid.
Antiviral Res. 88 : 287-295. - Bojkova D, Zoller N, Tietgen M, Steinhorst K, Bechtel M, Rothenburger T,
et al . 2023. Repurposing of the antibiotic nitroxoline for the treatment of mpox.J. Med. Virol. 95 : e28652. - Kindrachuk J, Arsenault R, Kusalik A, Kindrachuk KN, Trost B, Napper S,
et al . 2012. Systems kinomics demonstrates Congo Basin monkeypox virus infection selectively modulates host cell signaling responses as compared to West African monkeypox virus.Mol. Cell. Proteomics 11 : M111015701. - Beerli C, Yakimovich A, Kilcher S, Reynoso GV, Flaschner G, Muller DJ,
et al . 2019. Vaccinia virus hijacks EGFR signalling to enhance virus spread through rapid and directed infected cell motility.Nat. Microbiol. 4 : 216-225. - Hammarlund E, Lewis MW, Carter SV, Amanna I, Hansen SG, Strelow LI,
et al . 2005. Multiple diagnostic techniques identify previously vaccinated individuals with protective immunity against monkeypox.Nat. Med. 11 : 1005-1011. - Kemper AR, Davis MM, Freed GL. 2002. Expected adverse events in a mass smallpox vaccination campaign.
Eff. Clin. Pract. 5 : 84-90. - Edghill-Smith Y, Golding H, Manischewitz J, King LR, Scott D, Bray M,
et al . 2005. Smallpox vaccine-induced antibodies are necessary and sufficient for protection against monkeypox virus.Nat. Med. 11 : 740-747. - Edghill-Smith Y, Bray M, Whitehouse CA, Miller D, Mucker E, Manischewitz J,
et al . 2005. Smallpox vaccine does not protect macaques with AIDS from a lethal monkeypox virus challenge.J. Infect. Dis. 191 : 372-381. - Kenner J, Cameron F, Empig C, Jobes DV, Gurwith M. 2006. LC16m8: an attenuated smallpox vaccine.
Vaccine 24 : 7009-7022. - Maksyutov RA, Yakubitskiy SN, Kolosova IV, Tregubchak TV, Shvalov AN, Gavrilova EV,
et al . 2022. Genome stability of the vaccine strain VACDelta6.Vavilovskii Zhurnal Genet Selektsii. 26 : 394-401. - WHO. 2023. Mpox Vaccine Tracker: List of vaccine candidates in research & development. Available from https://www.who.int/publications/m/item/mpox-vaccine-tracker---list-of-vaccine-candidates-in-research---development. Accessed on April 23, 2023.
- Lum FM, Torres-Ruesta A, Tay MZ, Lin RTP, Lye DC, Renia L,
et al . 2022. Monkeypox: disease epidemiology, host immunity and clinical interventions.Nat. Rev. Immunol. 22 : 597-613. - Petersen BW, Harms TJ, Reynolds MG, Harrison LH. 2016. 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.
MMWR Morb. Mortal. Wkly. Rep. 65 : 257-262. - Pittman PR, Hahn M, Lee HS, Koca C, Samy N, Schmidt D,
et al . 2019. Phase 3 efficacy trial of modified vaccinia ankara as a vaccine against smallpox.N. Engl. J. Med. 381 : 1897-1908. - Thornhill JP, Barkati S, Walmsley S, Rockstroh J, Antinori A, Harrison LB,
et al . 2022. Monkeypox Virus Infection in Humans across 16 Countries - April-June 2022.N. Engl. J. Med. 387 : 679-691. - Rao AK, Petersen BW, Whitehill F, Razeq JH, Isaacs SN, Merchlinsky MJ,
et al . 2022. Use of JYNNEOS (Smallpox and Monkeypox Vaccine, Live, Nonreplicating) for Preexposure Vaccination of Persons at Risk for Occupational Exposure toOrthopoxviruses : Recommendations of the Advisory Committee on Immunization Practices - United States, 2022.MMWR Morb. Mortal. Wkly. Rep. 71 : 734-742. - Sang Y, Zhang Z, Liu F, Lu H, Yu C, Sun H,
et al . 2023. Monkeypox virus quadrivalent mRNA vaccine induces immune response and protects against vaccinia virus.Signal. Transduct. Target. Ther. 8 : 172. - Hooper JW, Custer DM, Schmaljohn CS, Schmaljohn AL. 2000. DNA vaccination with vaccinia virus L1R and A33R genes protects mice against a lethal poxvirus challenge.
Virology 266 : 329-339. - Fang Z, Monteiro VS, Renauer PA, Shang X, Suzuki K, Ling X,
et al . 2023. Polyvalent mRNA vaccination elicited potent immune response to monkeypox virus surface antigens.Cell. Res. 33 : 407-410. - Zeng J, Li Y, Jiang L, Luo L, Wang Y, Wang H,
et al . 2023. Mpox multi-antigen mRNA vaccine candidates by a simplified manufacturing strategy afford efficient protection against lethal orthopoxvirus challenge.Emerg. Microbes. Infect. 12 : 2204151. - Freyn AW, Atyeo C, Earl PL, Americo JL, Chuang G-Y, Natarajan H,
et al . 2022. A monkeypox mRNA-lipid nanoparticle vaccine targeting virus binding, entry, and transmission drives protection against lethal orthopoxviral challenge.bioRxiv. : 2022.2012.2017.520886. - Hou F, Zhang Y, Liu X, Murad Y, Xu J, Yu Z,
et al . 2022. Novel mRNA vaccines encoding Monkeypox virus M1R and A35R protect mice from a lethal virus challenge.bioRxiv. : 2022.2011.2019.517190. - Hooper JW, Thompson E, Wilhelmsen C, Zimmerman M, Ichou MA, Steffen SE,
et al . 2004. Smallpox DNA vaccine protects nonhuman primates against lethal monkeypox.J. Virol. 78 : 4433-4443. - Hooper JW, Custer DM, Thompson E. 2003. Four-gene-combination DNA vaccine protects mice against a lethal vaccinia virus challenge and elicits appropriate antibody responses in nonhuman primates.
Virology 306 : 181-195. - Mucker EM, Golden JW, Hammerbeck CD, Kishimori JM, Royals M, Joselyn MD,
et al . 2022. A nucleic acid-based orthopoxvirus vaccine targeting the vaccinia virus L1, A27, B5, and A33 proteins protects rabbits against lethal rabbitpox virus aerosol challenge.J. Virol. 96 : e0150421. - Heraud JM, Edghill-Smith Y, Ayala V, Kalisz I, Parrino J, Kalyanaraman VS,
et al . 2006. Subunit recombinant vaccine protects against monkeypox.J. Immunol. 177 : 2552-2564. - Hirao LA, Draghia-Akli R, Prigge JT, Yang M, Satishchandran A, Wu L,
et al . 2011. Multivalent smallpox DNA vaccine delivered by intradermal electroporation drives protective immunity in nonhuman primates against lethal monkeypox challenge.J. Infect. Dis. 203 : 95-102. - Franceschi V, Parker S, Jacca S, Crump RW, Doronin K, Hembrador E,
et al . 2015. BoHV-4-Based vector single heterologous antigen delivery protects STAT1(-/-) Mice from Monkeypoxvirus lethal challenge.PLoS Negl. Trop. Dis. 9 : e0003850. - Hernaez B, Alcami A. 2018. New insights into the immunomodulatory properties of poxvirus cytokine decoy receptors at the cell surface
7 : F1000Res. - Buchman GW, Cohen ME, Xiao Y, Richardson-Harman N, Silvera P, DeTolla LJ,
et al . 2010. A protein-based smallpox vaccine protects non-human primates from a lethal monkeypox virus challenge.Vaccine 28 : 6627-6636. - Xiao Y, Zeng Y, Schante C, Joshi SB, Buchman GW, Volkin DB,
et al . 2020. Short-term and longer-term protective immune responses generated by subunit vaccination with smallpox A33, B5, L1 or A27 proteins adjuvanted with aluminum hydroxide and CpG in mice challenged with vaccinia virus.Vaccine 38 : 6007-6018. - Patarroyo ME, Amador R, Clavijo P, Moreno A, Guzman F, Romero P,
et al . 1988. A synthetic vaccine protects humans against challenge with asexual blood stages of Plasmodium falciparum malaria.Nature 332 : 158-161. - Lozano JM, Rodriguez Parra Z, Hernandez-Martinez S, Yasnot-Acosta MF, Rojas AP, Marin-Waldo LS,
et al . 2021. The search of a Malaria vaccine: The time for modified immuno-potentiating probes.Vaccines (Basel) 9 : 115. - Lozano JM, Muller S. 2023. Monkeypox: potential vaccine development strategies.
Trends Pharmacol. Sci. 44 : 15-19. - Bonam SR, Partidos CD, Halmuthur SKM, Muller S. 2017. An overview of novel adjuvants designed for improving vaccine efficacy.
Trends Pharmacol. Sci. 38 : 771-793. - Blasco R, Moss B. 1991. Extracellular vaccinia virus formation and cell-to-cell virus transmission are prevented by deletion of the gene encoding the 37,000-Dalton outer envelope protein.
J. Virol. 65 : 5910-5920. - Fogg C, Lustig S, Whitbeck JC, Eisenberg RJ, Cohen GH, Moss B. 2004. Protective immunity to vaccinia virus induced by vaccination with multiple recombinant outer membrane proteins of intracellular and extracellular virions.
J. Virol. 78 : 10230-10237. - Ando J, Ngo MC, Ando M, Leen A, Rooney CM. 2020. Identification of protective T-cell antigens for smallpox vaccines.
Cytotherapy 22 : 642-652.