Skip to main content

Advancing the understanding and management of Mpox: insights into epidemiology, disease pathways, prevention, and therapeutic strategies

Abstract

Mpox, previously known as monkeypox, is a zoonotic viral disease caused by the Mpox virus (MPXV), a member of the Orthopoxvirus genus. This disease is of significant concern due to its zoonotic transmission, which can be challenging to control, its ability to spread easily from person to person, the potential for severe symptoms or even fatality, and its history of frequent global outbreaks. Despite the growing threat, there is still limited research on the pathophysiology of the disease and available disease-modifying treatments. To address this gap, the latest developments in Mpox epidemiology, viral variant detection, and advanced diagnostic tools for accurate MPXV detection have been reviewed. Ongoing preventive measures, including vaccination strategies, have also been examined. Additionally, the genomic and proteomic characteristics of MPXV have been explored, and network and pathway enrichment analyses have been performed to identify potential therapeutic targets. The findings presented in this manuscript suggest the potential for novel disease-modifying treatments. Moreover, emerging technologies, such as artificial intelligence and "big data," are playing a crucial role in advancing disease management and enhancing prevention strategies. This review emphasizes the evolving understanding of Mpox and MPXV variants and underscores the importance of continued research and public health initiatives to combat the disease and prevent future global outbreaks.

Peer Review reports

Introduction

Mpox, previously known as monkeypox, is a zoonotic disease caused by the Mpox virus (MPXV), a member of the Orthopoxvirus genus related to smallpox. It can be transmitted from animals to humans and between humans through direct contact, respiratory droplets, body fluids, or contaminated materials [1, 2]. The incubation period ranges from 1–21 days, with symptoms emerging within this timeframe and lasting 2–4 weeks. These include fever, headache, muscle aches, swollen lymph nodes, exhaustion, and a rash that evolves into pus-filled lesions, primarily on the face, hands, and feet. In severe cases, Mpox can be fatal [3]. The severity and duration of symptoms depend on various factors, including the patient's immune status and the viral strain [4,5,6]. One distinctive sign of MPXV infection is lymphadenectasis, which can help differentiate it from other Orthopoxvirus infections [7]. Mpox is endemic to West and Central Africa, though recent outbreaks have occurred in non-endemic regions such as Europe and North America.

Mpox is distinct from Monkey Fever, or Kyasanur Forest Disease (KFD), another zoonotic viral illness. KFD is caused by the Kyasanur Forest Disease virus (KFDV), a Flavivirus related to dengue, Zika, and yellow fever. It is transmitted to humans primarily through tick bites or direct contact with infected animals such as monkeys and rodents [8]. The incubation time ranges from 3–8 days and symptoms include fever, headache, and hemorrhagic manifestations (e.g., bleeding from gums, nose, and gastrointestinal tract in severe cases). In severe cases, Monkey Fever can be fatal. Unlike Mpox, which is more widespread, KFD is endemic to India. While both diseases are zoonotic, they differ in their causative agents, transmission, incubation time, symptoms, and geographic distribution [8].

Recent outbreaks of Mpox have garnered significant attention, driving increased interest in understanding the disease and developing effective treatments [6, 9,10,11,12,13,14,15]. Health agencies are particularly concerned for several reasons: the zoonotic nature of MPXV complicates control efforts, human-to-human transmission raises the risk of widespread outbreaks, and recent outbreaks from 2022 to 2024 have demonstrated the virus's ability to spread rapidly beyond endemic regions. Additionally, while symptoms are generally mild, they can become severe in immunocompromised individuals [16]. The absence of specific treatments for Mpox and the limited integration of vaccines into routine immunization programs further exacerbate the situation. Public awareness remains low, which can delay detection and increase risks for rapid global spread [16].

MPXV is categorized into two main clades: the Eurasian/African Orthopoxviruses and the North American Orthopoxviruses [16]. Specifically, clade I (subclades Ia and Ib) is endemic to Central and Eastern Africa, while clade II (subclades IIa and IIb) is primarily found in West Africa, where outbreaks are more common [1, 4, 17]. The 2022 Mpox outbreak, which began in West Africa, was caused by clade II and spread internationally, affecting over 100 countries across multiple continents [18, 19]. Clades I and IIa are zoonotic, whereas clade IIb primarily transmits between humans [20]. Transmission in endemic regions typically occurs through direct contact with infected animals during activities like hunting or processing animal products [21]. Small mammals and non-human primates can carry the virus asymptomatically [22], and various animal species, particularly rodents, serve as reservoirs for MPXV [23].

Human-to-human transmission can occur through sexual intercourse, as well as direct contact with respiratory droplets and bodily fluids [10, 24]. It can also occur through kitchen utensils, clothing, and bedding used by infected individuals [10]. MPXV transmission via direct inoculation has been reported in individuals after being pierced or having a tattoo on their skin, which both methods involve invasive techniques [25, 26]. Vertical transmission of MPXV can also happen from an infected mother to her fetus [10]. Furthermore, human-to-human transmission may occur through close or prolonged contact with infected individuals exhibiting disease symptoms, including face-to-face, skin-to-skin, mouth-to-mouth, or mouth-to-skin interactions [27, 28]. Healthcare workers are reported to have MPXV infection from infected patients in case protective personal equipment (PPE) were not worn [29]. The presence of MPXV in the air, specially from air samples collected around MPXV infected patients, along with patients’ exhaled droplets, may further play a role in MPXV transmission via air [30,31,32,33]. Although studies have not shown any link of human infection via aerosol transmission within the reported outbreaks [30], MPXV has been reported to be stable for up to 90 h in aerosols and might be a possible route for infection [29, 30, 34]. Several studies have demonstrated that aerosolized MPXV can be the source of infections in nonhuman primates [31, 35,36,37].

In contrast, animal-to-human transmission may happen via bites, scratches, direct contact with mucous membranes, body fluids, and tissues, or through the consumption of bush meat [10]. In addition, human-to-animal transmission has been reported in the recent outbreak in France in 2022 [38]. It was a transmission from a male to a dog, in which the dog was sharing a bed with two non-exclusive sexual partners males, and tests have shown that the virus in the dog and the one in one of the males were genetically identical [38, 39]. Additionally, MPXV may be transmitted between animals through direct contact between primates and rodents [18].

This overview will provide an update on the recent Mpox outbreaks comparing them with historical data to highlight trends in disease spread. Key topics to be covered include the latest diagnostic tools for MPXV detection, effective prevention strategies, available prevention methods including vaccines, and newly approved treatment options. A detailed exploration of the protein–protein interactions between MPXV-human hosts will be included, along with suggested potential treatment targets under investigation.

Mpox outbreaks and present situation

The history of MPXV can be tracked back to 1958 when it was first discovered in cynomolgus monkeys used for vaccine research purposes in Denmark [40]. Later, the first human Mpox case was reported in 1970 in Congo in a nine-month-old boy [4, 41]. The first human Mpox case in non-endemic areas was discovered in the US in 2003 [42]. After that, MPXV has been spreading throughout the world in several countries in various numbers until it became a risky outbreak recently. The World Health Organization (WHO) has declared Mpox a public health emergency of international concern (PHEIC) on July 23, 2022, since outbreaks of a high number of Mpox cases were confirmed in a range of countries that have not seen MPXV before [43]. PHEIC status for Mpox was later dropped on May 11, 2023, due to a decline in global cases [44], and later reinstated recently on August 14, 2024, due to an upsurge in Mpox cases in Congo and other neighboring African countries [45]. Figure 1 summarizes the timeline of Mpox outbreaks and major events from the time of MPXV discovery in 1958 until 2024 [6, 45].

Fig. 1
figure 1

The timeline of Mpox outbreaks and major events

As of January 1, 2024, a total of 125 countries have reported more than 9600 Mpox cases and more than 60 deaths [46]. These cases are divided as follows: 7 African countries have reported MPXV-clade I cases, 3 countries (in Africa, Europe, and Asia) have reported MPXV-clades I and II cases, 112 countries (in all continents) have reported MPXV-clade II cases, and 3 African countries have reported MPXV cases of unknown clade [46]. Furthermore, the recent outbreak is due to MPXV-clade IIb with 115 countries reporting MPXV cases for the first time [46]. The cumulative number of cases and deaths since June 2022 stands at more than 100,000 Mpox cases along with more than 200 deaths [47].

MPXV taxonomy, structure, and life cycle

MPXV belongs to the Orthopoxvirus genus within the Poxviridae family based on the most recent update from the International Committee on Taxonomy of Viruses [48]. The most recent Reference Sequence (RefSeq) for MPVX is NC_003310.1 by the National Center for Biotechnology Information (NCBI) [49]. MPXV is one of 13 species in the Orthopoxvirus genus, which are classified as enveloped viruses containing a single linear molecule of double-stranded DNA (dsDNA) [50]. Orthopoxvirus viruses are brick-shaped viruses with dimensions of about 200 × 200 × 250 nm with a characteristic dumbbell-shaped core [10], and a linear dsDNA comprising approximately 186–228 kbp that encodes between 174–233 proteins [50]. The core membrane, enclosing the core/nucleoprotein complex, is surrounded by a palisade layer, together forming what is known as the nucleosome complex [51].

Orthopoxvirus viruses also include the variola virus that causes smallpox disease and the vaccinia virus that causes cowpox disease [7]. As with all other enveloped viruses, MPXV’s infection and replication cycle involves 3 common steps: 1) attachment, entry, and uncoating; 2) DNA and protein synthesis; and 3) viral assembly, maturation, and release [52]. There are a few variations that occur for the MPXV’s replication cycle. Although MPXV is a DNA virus, once it enters the host cell, it will start replicating inside the cytoplasm, instead of inside the nucleus, using various encoded proteins instead of depending on the host cell's proteins [53, 54].

By the end of the replication cycle, two infectious forms of MPXV, i.e., virions, would be released and they include (Fig. 2): the intracellular mature virion (IMV) and the extracellular enveloped virion (EEV) [55]. IMV is an enveloped form of the virus, with its viral core surrounded by a single lipoprotein bilayer membrane [22]. In contrast, EEV is an enveloped form of the virus with an additional outer membrane compared to IMV [22]. IMVs initiate the infection, while EEVs have an extra outer membrane and are essential for evading host immune defenses and promoting viral spread among host species, emphasizing the complex lifecycle of MPXV [56, 57]. IMVs and EEVs can be released during host cell lysis [55], however, EEVs can also be released via exocytosis [21,22,23, 58]. IMVs enter the host cell through micropinocytosis, whereas EEV enters the host cell via fusion [21,22,23, 59]. The fusion process with the host cell membrane relies on several transmembrane proteins, while the stability of the IMVs or EEVs affects transmission between host animals and its spread within the host [60].

Fig. 2
figure 2

A schematic showing the enveloped viral particle of MPXV and key structural proteins. A) EEV virion. B) IMV virion

Due to its large size, MPXV encounters difficulties in efficiently evading host defenses and replicating speedily with an increased likelihood of triggering an immune response from the host [60]. To evade detection, MPXV has evolved a set of virulence genes that produce proteins capable of manipulating the host's immune response [61]. For example, the virotransducers, are intracellular proteins that reduce the cell response to viral infection. In contrast, the virostealth proteins hinder the visibility of immune system recognition bodies, such as cluster of differentiation 4 (CD4) and major histocompatibility complex 1 (MHC1). In addition, the extracellular viromimic proteins, including viroreceptors that bind to host cytokines and chemokines to disrupt their functions, and virokines, which mimic host cytokines, chemokines, and growth factors to promote viral replication and spread [60,61,62].

Once the virus enters the host cells, the virus replicates it DNA genome and transcribes mRNA using the host machinery, to produce viral proteins. Key proteins involved in the replication cycle include A29L, H3L, L1R/M1R, E8L, I5L, and A43R which will be discussed in Sect."MPXV Proteins". Protein synthesis is followed by viral assembly into IMVs, and some of these acquire an additional outer membrane to form EEVs. Both IMVs and EVVs can be released from infected cells during host cell lysis. However, EEVs can also be released through exocytosis, avoiding host immune detection.

MPXV genome and genomic viral variants

The genome size of MPXV is 196,858 bp with GC content of 33% according to NCBI’s reference genome sequence (RefSeq NC_003310.1) [49]. There are 10,560 viral sequences available on GISAID’s EpiPox [63], which is thus far the largest genomic database for depositing Mpox data. There are also 8,297 genome assemblies of MPXV genomes deposited at the Bacterial and Viral Bioinformatics Resource Center (BV-BRC) [64] and 8,297 genome assemblies deposited at NCBI [65]. An illustration of EpiPox genomic sequences by isolation geography (continent and country), and collection year is provided in Fig. 3A.

Fig. 3
figure 3

MPXV genomes and viral variants. A Pie charts for the genome frequencies based on isolation continent, isolation country and sample collection year. B Tracking global viral variants of MPXV from September 2023 through August 2024 with restricting the time window between September 2023 and July September 2024. These illustrations are based on Mpox GISAID’s data [66, 67]

Genomic analysis of MPXV samples by public health laboratories is essential for understanding global outbreaks and pandemic potential. Two distinct MPXV clades have been identified: clade I (with two subclades Ia and Ib) and clade II (with two subclades IIa and IIb), as mentioned above [4]. Clade I, found in the Congo Basin, is associated with up to 10% human mortality and is primarily transmitted by rodents, although human-to-human transmission can also occur, particularly in outbreaks [68]. Clade IIa, present in West Africa, causes a milder disease, has a low mortality rate, and is also a zoonotic infection. Clade IIb is a more recent and distinct genetic variant of the virus which is more commonly found in Central Africa and has been associated with more severe disease and higher mortality rates. The genetic factors underlying the differences in virulence and transmission are not yet fully understood due to the lack of appropriate small animal models that could meet the stringent safety requirements for studies [68].

Since November 2023, Congo has seen a significant increase in Mpox cases and the emergence of the new MPXV clade Ib variant. MPXV clade Ib, which was first identified in Congo has also been detected in Burundi, Kenya, Rwanda, Uganda, and other countries according to GISAID data [69]. Tracking global viral variants of MPXV from September 2023 through August 2024 is shown in Fig. 3B based on GISAID’s data.

MPXV proteins

The MPXV encodes a variety of proteins that play crucial roles in its replication, immune evasion, and pathogenesis. While the exact number of proteins may vary slightly depending on the viral strain, MPXV typically encodes around 200–250 proteins. These include structural proteins, non-structural proteins, and immunoregulatory proteins.

Structural proteins are essential for the formation of the virus particle (virion), its envelopment, assembly, and host-cell entry. These proteins make up the physical components of the virus, allowing it to infect new host cells. Structural proteins include those that form both the EEV and IMV as shown in Figs. 2 and 3. Three key structural proteins in the MPXV envelope are B6R, C19L, and A35R. Envelope protein B6R is crucial for viral particle assembly and may also play a role in immune evasion by interacting with the host's immune system. C19L is involved in immune modulation and may help the virus evade immune detection by interfering with host signaling pathways. A35R contributes to the stability of the virion and is important for ensuring the virus can exit host cells and infect other cells. The IMV is composed of additional key proteins, including A29L, H3L, L1R/M1R, E8L, I5L, and A43R, which play critical roles in the formation and stabilization of the virion.

On the other hand, non-structural proteins like A42R, A22R, E4L, E5R, F9R, H5R, and I3L are not part of the virion itself, but are vital for the virus’s replication cycle. Protein A42R plays a role in viral genome replication; A22R is part of the viral core and it is involved in the formation of viral replication machinery; E4L inhibits host responses by interfering with the interferon signaling pathway to promote viral replication; E5R enhances viral genome replication by interacting with host proteins; F9R facilitates the formation of the viral replication complex; H5R is involved in the regulation of the viral life cycle, likely interacting with cellular machinery to promote viral replication; and I3L plays a role in viral assembly and the in the overall viral replication cycle. These proteins play essential roles in immune evasion, host-cell manipulation, and viral replication and assembly. They are involved in processes such as viral transcription, translation, immune modulation, and cellular manipulation. Some key MPXV structural and non-structural proteins are depicted in Fig. 4 to indicate their corresponding gene locations on the viral genome.

Fig. 4
figure 4

A schematic representing the full length of the MPXV genome based on Reference Sequence (Ref. Seq) NC_003310.1. This schematic highlights important structural and non-structural proteins which play key roles in the viral life cycle and its infectivity

MPXV-host interactions

Mpox develops in two main disease stages after MPXV transmission to humans: an incubation stage lasting typically from 7 to 14 days, but can range from 5 to 21 days, where the infected individual is asymptomatic and non-contagious, followed by a prodromal stage, which is infectious [5, 70]. The incubation stage refers to the period between MPXV entry (through mucous membranes or damaged skin of the host) and the appearance of symptoms. During this time the virus is replicating at the infection site before spreading to regional lymph nodes and the bloodstream and causing primary viremia [21, 70, 71]. In animal-to-human transmission, lymph nodes are in fact the initial infection sites [5].

The prodromal stage occurs after the incubation stage, during which the patient experiences flu-like symptoms that signals the onset of infection. This phase lasts from 1 to 3 days and overlaps with the appearance of rash towards the end of this stage. During this stage the virus continues to spread through the blood stream and lymph nodes leading to secondary viremia and reaching distant organs including the skin. Symptoms like fever, chills, headache, muscle pain, fatigue, sore throat, and lymphadenopathy are prominent in this stage [4, 6, 72,73,74,75]. After the prodromal stage, the characteristic rash (often starting on the face and spreading to the body) appears as a hallmark rash of Mpox and it evolves into vesicles and pustules over 2–4 weeks [4, 6, 72,73,74,75] transitioning into pustules and scabs, which marks the start of the clinical stage. In this stage, MPXV deploys immune evasion strategies by binding to type I interferons, delaying immune activation, and suppressing inflammation, which aids in the spread of symptoms and disease [22, 71, 76].

There have been various atypical presentations of monkeypox virus (MPXV) infection reported in patients, with lesions appearing on different areas of the skin and mucous membranes [77]. For example, erythematous vesicles have been observed on hands, trunk, and legs [77]. Also, vesicles and ulcers have also been reported to appear on male genitals and perianal area [77]. Facial lesions, such as ulcers on the nose, cheeks, and upper lips, as well as bacterial infections in the pharynx and chin, have also been reported [77,78,79]. In some cases, atypical lesions at the same site may progress through different stages (such as vesicles, pustules, and crusts) rather than showing lesions at different stages across various body parts, which is the more typical pattern [80]. Other atypical presentations of MPXV infection include ulcers, papule on hand palm, and finger with acute paronychia and subungual ulcers [81].

MPXV immune evasion pathway

Orthopoxviruses are notable for their ability to evade the host immune system due to a set of genes that bypass pattern recognition receptors and disrupt the cellular apoptosis process [82,83,84]. In the human host, MPXV binds to type I interferons (IFN-alpha, IFN-beta, IFN-omega), delaying STAT1 activation and triggering apoptosis. It also prevents NF-κB nuclear translocation, suppressing inflammation-related genes, and binds to C3b and C4b to block complement activation. The MPXV-human host interactions leading to immune evasion and Mpox summarized in Fig. 5.

Fig. 5
figure 5

Mpox immune evasion pathway map. Nodes on the map correspond to network objects (genes, proteins, or chemicals). The R letter in the blue hexagon above each protein/gene indicates that the protein/gene is a drug target. The type of interaction or relation was reflected by an appropriate symbol placed in the middle of the link. B: binding; c; cleavage IE: influence on expression; TR: transcription regulation; red arrows for inhibition, green arrows for activation, violet arrows for emergent interaction due to disease, intermittent violet arrows for enhanced interaction, gray arrows for unspecified action, violet text boxes for normal process, pink text boxes for pathological processes, blue-lined white text boxes for notes. Map obtained from MetaCore.™ version 21.4 [85]

Pathways affected by immune evasion

Generating interactions networks using genes/protein that are part of the immune evasion pathway shown is Fig. 5, provides important insight into other potentially perturbed pathways in the human host in response to MPXV infection. Figure 6A shows a small interactions network of all genes/proteins in Mpox immune evasion pathway map from MetaCore™, while Fig. 6B shows an expanded interactions network resulting from adding 20 additional network seed nodes, which are nearest neighbor proteins to any of the proteins in smaller network (i.e., Fig. 6A). Enrichment analysis of the network nodes using Cytoscape [86] version 3.10.1. Network nodes in Fig. 5 are colored based on the top 10 enriched KEGG pathways.

Fig. 6
figure 6

Protein–protein interactions network of human host proteins impacted by MPXV. A Direct interactions network. B Expanded interactions network

We further conducted an enrichment analysis using MetaCore™ and included the top 10 enriched pathway maps in Table 1, alongside the top 10 KEGG pathways for direct interactions and expanded networks. This highlights pathways potentially affected by Mpox infection. While KEGG results lacked detail for understanding Mpox mechanisms, MetaCore™ provided greater mechanistic insight. Notably, the Mpox immune evasion pathway was the top enrichment result, reflecting our focus on the relevant genes/proteins. The second most significant finding was “glomerular injury in Lupus Nephritis,” suggesting Mpox may cause such injury, as confirmed by a recent report [87]. Additionally, two top results indicated the complement pathway's role in the host response to MPXV, with evidence showing MPXV is neutralized by complement-dependent antibodies in both infected and vaccinated individuals [88]. Two other top 10 pathways involved PKR, which plays a crucial role in the immune response by inhibiting viral replication through translation initiation blockade[89].

Table 1 Top enriched pathway maps using human host that are part of MPXV-host interactions network

Recent updates on Mpox diagnosis

The WHO currently recommends the use of polymerase chain reaction (PCR) testing for the diagnosis of Mpox, which involves identifying MPVX’s DNA [4]. Recent advancements and studies on the molecular diagnosis of Mpox along with PCR test are summarized in Table 2. It is important to note that there are variations in the expression of IgM and IgG antibodies following Mpox infection, particularly in terms of their timing and duration. IgM antibodies typically appear with the onset of the rash, peak after two weeks, and decline within a year [23, 90]. In contrast, IgG antibodies also rise rapidly at the onset of the rash, peak after six weeks, and can persist in the body for decades [23, 90].

Table 2 Updated list of Mpox diagnostics methods

Based on the recent outbreaks of Mpox and its presence all around the world, there is an urgent need to advance diagnostic capabilities. Ensuring that all countries have access to affordable and accessible diagnostic tests is crucial, regardless of their economic capacity or healthcare infrastructure. Such advancements would significantly enhance efforts to diagnose patients accurately, identify the circulating viral strains, and contribute to the eventual eradication of the disease [91].

Prevention, vaccination, and other treatment options for Mpox

The integration of public health strategies, vaccination programs, and antiviral research is essential for controlling Mpox outbreaks and protecting public health. These strategies are discussed in the following subsections.

Prevention

To prevent the transmission of Mpox between animals and humans and among humans, several measures can be implemented [117]. These measures aim to control both primary transmission (animal-to-human) and secondary transmission (human-to-human). Exposed pet animals should be isolated from other animals and humans in designated areas, while Mpox patients must also be isolated until fully recovered. Specific precautions should be taken in various settings, including healthcare facilities and airports, such as wearing PPE, maintaining physical distance, and using disinfectants [29]. The WHO recommends that infected patients remain at home in a well-ventilated room and avoid contact with others [4]. Other self-care and prevention methods as outlined by the WHO include [4]: if patients must be around others, they should cover any skin lesions and wear masks; patients should wash their hands thoroughly after touching sores, disinfect shared items, and can use saltwater rinses for mouth sores and warm baths with baking soda or Epsom salts for body sores; over-the-counter pain relief can be administered; and avoid actions that could spread the rash, such as popping blisters or shaving affected areas.

Vaccines

Mpox is a vaccine-preventable disease, similar to influenza, polio, cholera, and COVID- 19 [118,119,120,121,122], but vaccination is not part of mandatory national programs. It is recommended for high-risk individuals (pre-exposure) and those exposed to the virus (post-exposure), ideally within 4 days of contact or 14 days if the individual is asymptomatic [4]. Mpox vaccines (Table 3) can prevent the disease and generate sufficient antibodies, although efficacy may decline over time and vary by dose [123].

Table 3 FDA approved vaccines that can be used to protect from Mpox

Vaccination against Mpox reduces transmission, controls outbreaks, and lessens disease severity. While mass vaccination is not currently recommended due to the assessed risks and benefits, targeted vaccination campaigns combined with behavioral changes in high-risk groups have been effective in reducing the spread of Mpox [100, 123]. A study of the 2022–2023 US outbreak showed high transmission rates among high-risk populations, especially men with male-to-male sexual contact. The vaccination campaign, covering 37% of the high-risk population, prevented 21.2% of cases, while combining vaccines with behavior changes prevented up to 64% of cases, leading to a significant flattening of the epidemic curve [131]. These findings highlight the effectiveness of integrated prevention strategies in reducing the disease burden during outbreaks.

Furthermore, a closer look at adverse events following immunization (AEFI) data from the Netherlands and the World Health Organization highlighted a range of reactions, primarily focused on injection site issues and typical systemic responses. These reactions, while generally mild, offer valuable insights into the safety profile of the Mpox vaccines and help to inform both public health strategies and individual decisions regarding vaccination [129].

Small-molecule antiviral drugs

Several antiviral drugs have been investigated for the treatment of Mpox, as outlined in Table 4. Numerous studies have evaluated their effectiveness in managing the disease.

Table 4 Approved small-molecule chemical antiviral drugs for Mpox and examples of ongoing clinical trials

Drugs and vaccines under active development for Mpox

There are several drugs that are currently being under active development for the treatment and/or prevention of Mpox, according to the Cortellis Drug Discovery Intelligence (CDDI) database [151], These drugs are listed in Table 5.

Table 5 Drugs under active development for Mpox and/or other Orthopoxviruses

Suggested targets for therapeutic intervention and drug repurposing based on network pharmacology

In the quest for effective treatments for Mpox, identifying viral and host proteins that facilitate virus-host interactions is crucial. These proteins play essential roles in the virus's life cycle, making them promising targets for drug discovery. By focusing on these putative targets, researchers can develop novel therapeutic strategies aimed at disrupting the mechanisms through which the virus replicates and spreads. To aid in this effort, we have summarized relevant genes and proteins in Table 6 which provides a comprehensive overview of the key viral and host factors involved in Mpox pathogenesis. This table highlights the potential significance of identified drug targets in therapeutic interventions to guide future research directions. We also hypothesize that drugs targeting protein targets listed in Table 6 can be repurposed for combating Mpox and should be studied thoroughly by the scientific community.

Table 6 MPXV and human host genes/proteins suggested as therapeutic targets for Mpox based on network analyses described in work

Updates on Mpox Resources and Innovative Solutions

The integration of innovative resources such as genomic databases, artificial intelligence applications, data-sharing initiatives, and various open-source educational tools plays a crucial role in enhancing our understanding and management of Mpox. Table 7 provides and up-to-date list of resources available for researchers and healthcare professionals.

Table 7 List of popular innovative Mpox resources and databases

Conclusions

In conclusion, the evolving landscape of Mpox presents significant challenges and opportunities for public health and drug discovery research. As this zoonotic disease continues to spread internationally, understanding its epidemiological trends, transmission dynamics, viral variants, and disease pathways becomes increasingly critical to develop preventive measures and specific disease-modifying therapeutics. This updated review has highlighted the importance of early detection and innovative diagnostic tools, alongside effective preventive measures, such as available vaccines and small-molecule therapeutic agents. In addition, ongoing research into disease pathways, including viral-host interactions and immune response mechanisms, is essential for the development of targeted therapies. The genomic analyses, network pharmacology, and enriched pathway studies highlighted critical interactions between MPXV and host immune responses, informing future drug discovery research and public health strategies. Furthermore, the integration of advanced technologies, including AI and big data analytics, is hoped to revolutionize viral variant tracking, disease management, and response efforts. As we move forward, it is imperative that we remain vigilant and proactive in addressing this emerging public health threat, continually adapting our strategies to new developments in the prevention and treatment of Mpox.

Data availability

All data are presented within the manuscript. Supporting data files for the network and enrichment results can be accessed on GitHub at https://github.com/rhajjo/Mpox.

Abbreviations

bp:

Base pairs

CDC:

Centers for Disease Control and Prevention

CD4:

Cluster of differentiation 4

CDDI:

Cortellis Drug Discovery Intelligence

CP:

Core protein

CRISPR:

Clustered regularly interspaced short palindromic repeats

DRC:

Democratic Republic of the Congo

EEV:

Extracellular enveloped virion

ELISA:

Enzyme‐linked immunosorbent assay

FDA:

US Food and Drug Administration

GISAID:

Global Initiative on Sharing All Influenza Data

IMV:

Intracellular mature virion

IV:

Intravenous

MC:

Molluscum contagiosum

MCV:

Molluscum contagiosum virus

MHC1:

Major histocompatibility complex 1

MPXV:

Monkeypox virus

MC:

Molluscum contagiosum

NCBI:

National Center for Biotechnology Information

ORF:

Open reading frame

PHEIC:

Public health emergency of international concern

PCR:

Polymerase chain reaction

RT-PCR:

Real-time polymerase chain reaction

RPA:

Recombinase polymerase amplification

TEM:

Transmission electron microscopy

WHO:

World Health Organization

References

  1. CDC | About Mpox; available from: https://www.cdc.gov/mpox/about/?CDC_AAref_Val=https://www.cdc.gov/poxvirus/mpox/about/index.html. Accessed 10 Sept 2024.

  2. WHO | WHO recommends new name for monkeypox disease; available from: https://www.who.int/news/item/28-11-2022-who-recommends-new-name-for-monkeypox-disease. Accessed 10 Sept 2024.

  3. Reynolds MG, McCollum AM, Nguete B, Shongo Lushima R, Petersen BW. Improving the Care and Treatment of Monkeypox Patients in Low–Resource Settings: Applying Evidence from Contemporary Biomedical and Smallpox Biodefense Research. Viruses. 2017;9(12):380.

    Article  PubMed  PubMed Central  Google Scholar 

  4. WHO | Mpox; available from: https://www.who.int/news-room/fact-sheets/detail/mpox. Accessed 10 Sept 2024.

  5. Kaler J, Hussain A, Flores G, Kheiri S, Desrosiers D. Monkeypox: A Comprehensive Review of Transmission, Pathogenesis, and Manifestation. Cureus. 2022;14(7):e26531.

    PubMed  PubMed Central  Google Scholar 

  6. Lu J, Xing H, Wang C, Tang M, Wu C, Ye F, Yin L, Yang Y, Tan W, Shen L. Mpox (formerly monkeypox): pathogenesis, prevention, and treatment. Signal Transduct Target Ther. 2023;8(1):458.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shafaati M, Zandi M. State-of-the-art on monkeypox virus: an emerging zoonotic disease. Infection. 2022;50(6):1425–1430.

    Article  PubMed  Google Scholar 

  8. N S, Kandi V, G SR, Ca J, A H, As A, Kapil C, Palacholla PS: Kyasanur Forest Disease: A Comprehensive Review. Cureus. 2024;16(7):e65228.

  9. Shamim MA, Satapathy P, Padhi BK, Veeramachaneni SD, Akhtar N, Pradhan A, Agrawal A, Dwivedi P, Mohanty A, Pradhan KB, et al. Pharmacological treatment and vaccines in monkeypox virus: a narrative review and bibliometric analysis. Front Pharmacol. 2023;14:1149909.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Karagoz A, Tombuloglu H, Alsaeed M, Tombuloglu G, AlRubaish AA, Mahmoud A, Smajlović S, Ćordić S, Rabaan AA, Alsuhaimi E. Monkeypox (mpox) virus: Classification, origin, transmission, genome organization, antiviral drugs, and molecular diagnosis. J Infect Public Health. 2023;16(4):531–41.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Rabaan AA, Abas AH, Tallei TE, Al-Zaher MA, Al-Sheef NM, Fatimawali, Al-Nass EZ, Al-Ebrahim EA, Effendi Y, Idroes R, et al: Monkeypox outbreak 2022: What we know so far and its potential drug targets and management strategies. J Med Virol. 2023;95(1):e28306.

  12. Soheili M, Nasseri S, Afraie M, Khateri S, Moradi Y, Mahdavi Mortazavi SM, Gilzad-Kohan H. Monkeypox: Virology, Pathophysiology, Clinical Characteristics, Epidemiology, Vaccines, Diagnosis, and Treatments. J Pharm Pharm Sci. 2022;25:297–322.

    Article  CAS  PubMed  Google Scholar 

  13. Bruno G, Buccoliero GB. Antivirals against Monkeypox (Mpox) in Humans: An Updated Narrative Review. Life. Basel: 2023;13(10):1969.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Rizk JG, Lippi G, Henry BM, Forthal DN, Rizk Y. Prevention and Treatment of Monkeypox. Drugs. 2022;82(9):957–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Saied AA, Dhawan M, Metwally AA, Fahrni ML, Choudhary P, Choudhary OP. Disease History, Pathogenesis, Diagnostics, and Therapeutics for Human Monkeypox Disease: A Comprehensive Review. Vaccines. 2022;10(12):2091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Emerson GL, Li Y, Frace MA, Olsen-Rasmussen MA, Khristova ML, Govil D, Sammons SA, Regnery RL, Karem KL, Damon IK, et al. The phylogenetics and ecology of the orthopoxviruses endemic to North America. PLoS ONE. 2009;4(10):e7666.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Scarpa F, Sanna D, Azzena I, Cossu P, Locci C, Angeletti S, Maruotti A, Ceccarelli G, Casu M, Fiori PL, et al. Genetic Variability of the Monkeypox Virus Clade IIb B.1. J Clin Med. 2022;11(21):6388.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bunge EM, Hoet B, Chen L, Lienert F, Weidenthaler H, Baer LR, Steffen R. The changing epidemiology of human monkeypox-A potential threat? A systematic review. PLoS Negl Trop Dis. 2022;16(2):e0010141.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Meo SA, Al-Khlaiwi T, Al Jassir FF, Meo AS. Impact of traveling on transmission trends of human monkeypox disease: worldwide data based observational analysis. Front Public Health. 2023;11:1029215.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Andrei G, Snoeck R. Differences in pathogenicity among the mpox virus clades: impact on drug discovery and vaccine development. Trends Pharmacol Sci. 2023;44(10):719–39.

    Article  CAS  PubMed  Google Scholar 

  21. Kumar N, Acharya A, Gendelman HE, Byrareddy SN. The 2022 outbreak and the pathobiology of the monkeypox virus. J Autoimmun. 2022;131:102855.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Martínez-Fernández DE, Fernández-Quezada D, Casillas-Muñoz FAG, Carrillo-Ballesteros FJ, Ortega-Prieto AM, Jimenez-Guardeño JM, Regla-Nava JA. Human Monkeypox: A Comprehensive Overview of Epidemiology, Pathogenesis, Diagnosis, Treatment, and Prevention Strategies. Pathog. 2023;12(7):947.

    Article  Google Scholar 

  23. Huang Y, Mu L, Wang W. Monkeypox: epidemiology, pathogenesis, treatment and prevention. Signal Transduct Target Ther. 2022;7(1):373.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Wannigama DL, Amarasiri M, Phattharapornjaroen P, Hurst C, Modchang C, Besa JJV, Miyanaga K, Cui L, Fernandez S, Huang AT, et al. Community-based mpox and sexually transmitted disease surveillance using discarded condoms in the global south. Lancet Infect Dis. 2024;24(10):e610–3.

    Article  PubMed  Google Scholar 

  25. Viedma-Martinez M, Dominguez-Tosso FR, Jimenez-Gallo D, Garcia-Palacios J, Riera-Tur L, Montiel-Quezel N, Linares-Barrios M. MPXV Transmission at a Tattoo Parlor. N Engl J Med. 2023;388(1):92–4.

    Article  PubMed  Google Scholar 

  26. Tascini C, Geminiani M, Sbrana F, Pagotto A, Martini L. Possible tattoo-transmitted monkeypox viral infection. Intern Emerg Med. 2022;17(8):2421–2.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Gaspari V, Rossini G, Robuffo S, Rapparini L, Scagliarini A. Mistral De Pascali A, Piraccini BM, Lazzarotto T: Monkeypox Outbreak 2022: Clinical and Virological Features of 30 Patients at the Sexually Transmitted Diseases Centre of Sant’ Orsola Hospital, Bologna, Northeastern Italy. J Clin Microbiol. 2023;61(1):e0136522.

    Article  PubMed  Google Scholar 

  28. Wang Y, Leng P, Zhou H. Global transmission of monkeypox virus-a potential threat under the COVID-19 pandemic. Front Immunol. 2023;14:1174223.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Saied AA, Chandran D, Chakraborty S, Emran TB, Dhama K. Mpox and healthcare workers — a minireview of our present knowledge. The Egyptian Journal of Internal Medicine. 2023;35(1):46.

    Article  Google Scholar 

  30. Saied AA. Should not airborne transmission be ignored in the 2022 monkeypox outbreak? Int J Surg. 2022;104:106762.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Saied AA. Mpox virus Clade IIb detection in the air. J Med Virol. 2023;95(5):e28775.

    Article  CAS  PubMed  Google Scholar 

  32. Gould S, Atkinson B, Onianwa O, Spencer A, Furneaux J, Grieves J, Taylor C, Milligan I, Bennett A, Fletcher T, et al. Air and surface sampling for monkeypox virus in a UK hospital: an observational study. Lancet Microbe. 2022;3(12):e904–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hernaez B, Muñoz-Gómez A, Sanchiz A, Orviz E, Valls-Carbo A, Sagastagoitia I, Ayerdi O, Martín R, Puerta T, Vera M, et al. Monitoring monkeypox virus in saliva and air samples in Spain: a cross-sectional study. Lancet Microbe. 2023;4(1):e21–8.

    Article  CAS  PubMed  Google Scholar 

  34. Verreault D, Killeen SZ, Redmann RK, Roy CJ. Susceptibility of monkeypox virus aerosol suspensions in a rotating chamber. J Virol Methods. 2013;187(2):333–7.

    Article  CAS  PubMed  Google Scholar 

  35. Barnewall RE, Fisher DA, Robertson AB, Vales PA, Knostman KA, Bigger JE. Inhalational monkeypox virus infection in cynomolgus macaques. Front Cell Infect Microbiol. 2012;2:117.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Nalca A, Livingston VA, Garza NL, Zumbrun EE, Frick OM, Chapman JL, Hartings JM. Experimental infection of cynomolgus macaques (Macaca fascicularis) with aerosolized monkeypox virus. PLoS One. 2010;5(9):e12880.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Zaucha GM, Jahrling PB, Geisbert TW, Swearengen JR, Hensley L. The pathology of experimental aerosolized monkeypox virus infection in cynomolgus monkeys (Macaca fascicularis). Lab Invest. 2001;81(12):1581–600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Seang S, Burrel S, Todesco E, Leducq V, Monsel G, Le Pluart D, Cordevant C, Pourcher V, Palich R. Evidence of human-to-dog transmission of monkeypox virus. The Lancet. 2022;400(10353):658–9.

    Article  Google Scholar 

  39. Choudhary OP, Priyanka, Chopra H, Shafaati M, Dhawan M, Metwally AA, Saied AA, Rabaan AA, Alhumaid S, Al Mutair A, et al. Reverse zoonosis and its relevance to the monkeypox outbreak 2022. New Microbes New Infect. 2022;49–50:101049.

  40. Pv M. Andersen EK, Petersen KB, Birch-Andersen A: A POX-LIKE DISEASE IN CYNOMOLGUS MONKEYS. Acta Pathologica Microbiologica Scandinavica. 1959;46(2):156–76.

    Article  Google Scholar 

  41. Breman JG, Kalisa R, Steniowski MV, Zanotto E, Gromyko AI, Arita I. Human monkeypox, 1970–79. Bull World Health Organ. 1980;58(2):165–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Reed KD, Melski JW, Graham MB, Regnery RL, Sotir MJ, Wegner MV, Kazmierczak JJ, Stratman EJ, Li Y, Fairley JA, et al. The detection of monkeypox in humans in the Western Hemisphere. N Engl J Med. 2004;350(4):342–50.

    Article  CAS  PubMed  Google Scholar 

  43. WHO | WHO Director-General's statement at the press conference following IHR Emergency Committee regarding the multi-country outbreak of monkeypox - 23 July 2022; available from: https://www.who.int/director-general/speeches/detail/who-director-general-s-statement-on-the-press-conference-following-IHR-emergency-committee-regarding-the-multi--country-outbreak-of-monkeypox--23-july-2022. Accessed 13 Sept 2024.

  44. WHO | Fifth Meeting of the International Health Regulations (2005) (IHR) Emergency Committee on the Multi-Country Outbreak of mpox (monkeypox); available from: https://www.who.int/news/item/11-05-2023-fifth-meeting-of-the-international-health-regulations-(2005)-(ihr)-emergency-committee-on-the-multi-country-outbreak-of-monkeypox-(mpox). Accessed 13 Sept 2024.

  45. WHO | WHO Director-General declares mpox outbreak a public health emergency of international concern; available from: https://www.who.int/news/item/14-08-2024-who-director-general-declares-mpox-outbreak-a-public-health-emergency-of-international-concern. Accessed 10 Sept 2024).

  46. CDC | Mpox in the United States and Around the World: Current Situation Summary; available from: https://www.cdc.gov/poxvirus/mpox/outbreak/index.html. Accessed 10 Sept 2024.

  47. WHO | 2022–24 Mpox (Monkeypox) Outbreak: Global Trends; available from: https://worldhealthorg.shinyapps.io/mpx_global/#34_Tables. Accessed 10 Sept 2024.

  48. ICTV | Taxon Details: Orthopoxvirus monkeypox; available from: https://ictv.global/taxonomy/taxondetails?taxnode_id=202304771&taxon_name=Orthopoxvirus%20monkeypox. Accessed 10 Sept 2024.

  49. NCBI |Genome assembly ViralProj15142; available from: https://www.ncbi.nlm.nih.gov/datasets/genome/GCF_000857045.1/. Accessed 19 Sept 2024.

  50. ICTV | Genus: Orthopoxvirus; available from: https://ictv.global/report/chapter/poxviridae/poxviridae/orthopoxvirus. Accessed 10 Sept 2024.

  51. Wang Y, Yang K, Zhou H. Immunogenic proteins and potential delivery platforms for mpox virus vaccine development: A rapid review. Int J Biol Macromol. 2023;245:125515.

    Article  CAS  PubMed  Google Scholar 

  52. Hajjo R, Sabbah DA, Abusara OH, Kharmah R, Bardaweel S. Targeting Human Proteins for Antiviral Drug Discovery and Repurposing Efforts: A Focus on Protein Kinases. Viruses. 2023;15(2):568.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Buller RM, Palumbo GJ. Poxvirus pathogenesis. Microbiol Rev. 1991;55(1):80–122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Stanford MM, McFadden G, Karupiah G, Chaudhri G. Immunopathogenesis of poxvirus infections: forecasting the impending storm. Immunol Cell Biol. 2007;85(2):93–102.

    Article  CAS  PubMed  Google Scholar 

  55. Lansiaux E, Jain N, Laivacuma S, Reinis A. The virology of human monkeypox virus (hMPXV): A brief overview. Virus Res. 2022;322:198932.

    Article  CAS  PubMed  Google Scholar 

  56. Tolonen N, Doglio L, Schleich S, Krijnse Locker J. Vaccinia virus DNA replication occurs in endoplasmic reticulum-enclosed cytoplasmic mini-nuclei. Mol Biol Cell. 2001;12(7):2031–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Moss B. Poxvirus cell entry: how many proteins does it take? Viruses. 2012;4(5):688–707.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Gong Q, Wang C, Chuai X, Chiu S. Monkeypox virus: a re-emergent threat to humans. Virol Sin. 2022;37(4):477–82.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Moss B. Membrane fusion during poxvirus entry. Semin Cell Dev Biol. 2016;60:89–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kieser Q, Noyce RS, Shenouda M, Lin YJ, Evans DH. Cytoplasmic factories, virus assembly, and DNA replication kinetics collectively constrain the formation of poxvirus recombinants. PLoS ONE. 2020;15(1):e0228028.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Petersen E, Kantele A, Koopmans M, Asogun D, Yinka-Ogunleye A, Ihekweazu C, Zumla A. Human Monkeypox: Epidemiologic and Clinical Characteristics, Diagnosis, and Prevention. Infect Dis Clin North Am. 2019;33(4):1027–43.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Chen N, Li G, Liszewski MK, Atkinson JP, Jahrling PB, Feng Z, Schriewer J, Buck C, Wang C, Lefkowitz EJ, et al. Virulence differences between monkeypox virus isolates from West Africa and the Congo basin. Virology. 2005;340(1):46–63.

    Article  CAS  PubMed  Google Scholar 

  63. Ballesteros-Sanabria L, Pelaez-Prestel HF, Reche PA, Lafuente EM. EPIPOX: A Resource Facilitating Epitope-Vaccine Design Against Human Pathogenic Orthopoxviruses. Methods Mol Biol. 2023;2673:175–85.

    Article  CAS  PubMed  Google Scholar 

  64. BV-BRC | Viruses: Genomes; available from: https://www.bv-brc.org/view/Taxonomy/10244. Accessed 19 Sept 2024.

  65. NCBI | Genome: Monkeypox virus; available from: https://www.ncbi.nlm.nih.gov/datasets/genome/?taxon=10244. Accessed 19 Sept 2024.

  66. Elbe S, Buckland-Merrett G. Data, disease and diplomacy: GISAID’s innovative contribution to global health. Glob Chall. 2017;1(1):33–46.

    Article  PubMed  PubMed Central  Google Scholar 

  67. GISAID | GISAID Database; available from: https://gisaid.org/. Accessed 23 Sept 2024.

  68. Americo JL, Earl PL, Moss B: Virulence differences of mpox (monkeypox) virus clades I, IIa, and IIb.1 in a small animal model. Proc Natl Acad Sci U S A. 2023;120(8):e2220415120.

  69. GISAID | Tracking of Mpox Variants: Clade Ib; available from: https://gisaid.org/mpox-variants/. Accessed 15 Sept 2024.

  70. Moore MJ, Rathish B, Zahra F: Mpox (Monkeypox). In: StatPearls. edn. Treasure Island (FL): StatPearls Publishing Copyright © 2024, StatPearls Publishing LLC.; 2024.

  71. Alakunle E, Moens U, Nchinda G, Okeke MI. Monkeypox Virus in Nigeria: Infection Biology, Epidemiology, and Evolution. Viruses. 2020;12(11):1257.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Walter K, Malani PN. What Is Monkeypox? JAMA. 2022;328(2):222.

    Article  PubMed  Google Scholar 

  73. Cheema AY, Ogedegbe OJ, Munir M, Alugba G, Ojo TK. Monkeypox: A Review of Clinical Features, Diagnosis, and Treatment. Cureus. 2022;14(7):e26756.

    PubMed  PubMed Central  Google Scholar 

  74. Pittman PR, Martin JW, Kingebeni PM, Tamfum J-JM, Wan Q, Reynolds MG, Quinn X, Norris S, Townsend MB, Satheshkumar PS, et al. Clinical characterization of human monkeypox infections in the Democratic Republic of the Congo. medRxiv. 2022:2022.2005.2026.22273379.

  75. Benites-Zapata VA, Ulloque-Badaracco JR, Alarcon-Braga EA, Hernandez-Bustamante EA, Mosquera-Rojas MD, Bonilla-Aldana DK, Rodriguez-Morales AJ. Clinical features, hospitalisation and deaths associated with monkeypox: a systematic review and meta-analysis. Ann Clin Microbiol Antimicrob. 2022;21(1):36.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Malik S, Ahmed A, Ahsan O, Muhammad K, Waheed Y. Monkeypox Virus: A Comprehensive Overview of Viral Pathology, Immune Response, and Antiviral Strategies. Vaccines (Basel). 2023;11(8):1345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Ciccarese G, Di Biagio A, Bruzzone B, Guadagno A, Taramasso L, Oddenino G, Brucci G, Labate L, De Pace V, Mastrolonardo M, et al. Monkeypox outbreak in Genoa, Italy: Clinical, laboratory, histopathologic features, management, and outcome of the infected patients. J Med Virol. 2023;95(2):e28560.

    Article  CAS  PubMed  Google Scholar 

  78. Wick JM, Pelliccione A, Lee-Rodriguez C, Tran HN, McCleskey PE, Nichol A, Skarbinski J. An Atypical Presentation of Mpox Transmitted Between Transgender Men Through Oral Sex. Sex Transm Dis. 2023;50(10):685–6.

    PubMed  Google Scholar 

  79. Elsheikh N, Salvia Milos A. Atypical Clinical presentation of a patient with Monkeypox. Ir Med J. 2023;116(5):770.

    CAS  PubMed  Google Scholar 

  80. Pérez-Martín ÓG, Hernández-Aceituno A, Dorta-Espiñeira MM, García-Hernández L, Larumbe-Zabala E. Atypical presentation of sexually-transmitted monkeypox lesions. Infect Dis (Lond). 2022;54(12):940–3.

    Article  PubMed  Google Scholar 

  81. Frade JV, Sousa I, Marques T, Filipe P. Acute paronychia: An atypical presentation of Monkeypox infection. Enferm Infecc Microbiol Clin. 2023;41(5):317–8.

    Article  PubMed  Google Scholar 

  82. Zandi M, Shafaati M, Hosseini F. Mechanisms of immune evasion of monkeypox virus. Front Microbiol. 2023;14:1106247.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Li H, Huang QZ, Zhang H, Liu ZX, Chen XH, Ye LL, Luo Y. The land-scape of immune response to monkeypox virus. EBioMedicine. 2023;87:104424.

    Article  CAS  PubMed  Google Scholar 

  84. Yu H, Bruneau RC, Brennan G, Rothenburg S. Battle Royale: Innate Recognition of Poxviruses and Viral Immune Evasion. Biomedicines. 2021;9(7):765.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Clarivate | Metacore - Integrated pathway analysis for multi-OMICs data; https://clarivate.com/life-sciences-healthcare/webinars/metacore-integrated-pathway-analysis-for-multi-omics-data/. Accessed 10 Sept 2024.

  86. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Reis T, de Assis Rocha Neves F, Fagundes A, Jr., See E, da Hora Passos R, Zawadzki B, Bellomo R, Ronco C: Monkeypox-Associated Acute Kidney Injury and Foreseeable Impacts on Nephrology and Kidney Transplantation Services. Blood Purification .2023;52(4):319–322.

  88. Hubert M, Guivel-Benhassine F, Bruel T, Porrot F, Planas D, Vanhomwegen J, Wiedemann A, Burrel S, Marot S, Palich R, et al. Complement-dependent mpox-virus-neutralizing antibodies in infected and vaccinated individuals. Cell Host Microbe. 2023;31(6):937-948.e934.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Langland JO, Jacobs BL. Inhibition of PKR by vaccinia virus: role of the N- and C-terminal domains of E3L. Virology. 2004;324(2):419–29.

    Article  CAS  PubMed  Google Scholar 

  90. Karem KL, Reynolds M, Braden Z, Lou G, Bernard N, Patton J, Damon IK. 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. 2005;12(7):867–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Lim CK, Roberts J, Moso M, Liew KC, Taouk ML, Williams E, Tran T, Steinig E, Caly L, Williamson DA. Mpox diagnostics: Review of current and emerging technologies. J Med Virol. 2023;95(1):e28429.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Realegeno S, Puschnik AS, Kumar A, Goldsmith C, Burgado J, Sambhara S, Olson VA, Carroll D, Damon I, Hirata T, et al. Monkeypox Virus Host Factor Screen Using Haploid Cells Identifies Essential Role of GARP Complex in Extracellular Virus Formation. J Virol. 2017;91(11):e00011-17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Cho CT, Wenner HA. In vitro growth characteristics of monkeypox virus. Proc Soc Exp Biol Med. 1972;139(3):916–20.

    Article  CAS  PubMed  Google Scholar 

  94. Gelderblom HR, Hazelton PR. Specimen collection for electron microscopy. Emerg Infect Dis. 2000;6(4):433–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Bayer-Garner IB. Monkeypox virus: histologic, immunohistochemical and electron-microscopic findings. J Cutan Pathol. 2005;32(1):28–34.

    Article  CAS  PubMed  Google Scholar 

  96. Stagles MJ, Watson AA, Boyd JF, More IA, McSeveney D. The histopathology and electron microscopy of a human monkeypox lesion. Trans R Soc Trop Med Hyg. 1985;79(2):192–202.

    Article  CAS  PubMed  Google Scholar 

  97. Esposito JJ, Obijeski JF, Nakano JH. Serological relatedness of monkeypox, variola, and vaccinia viruses. J Med Virol. 1977;1(1):35–47.

    Article  CAS  PubMed  Google Scholar 

  98. Nalca A, Rimoin AW, Bavari S, Whitehouse CA. Reemergence of monkeypox: prevalence, diagnostics, and countermeasures. Clin Infect Dis. 2005;41(12):1765–71.

    Article  PubMed  Google Scholar 

  99. Gispen R, Huisman J, Brand-Saathof B, Hekker AC. Immunofluorescence test for persistent poxvirus antibodies. Arch Gesamte Virusforsch. 1974;44(4):391–5.

    Article  CAS  PubMed  Google Scholar 

  100. Hammarlund E, Lewis MW, Carter SV, Amanna I, Hansen SG, Strelow LI, Wong SW, Yoshihara P, Hanifin JM, Slifka MK. Multiple diagnostic techniques identify previously vaccinated individuals with protective immunity against monkeypox. Nat Med. 2005;11(9):1005–11.

    Article  CAS  PubMed  Google Scholar 

  101. Dubois ME, Hammarlund E, Slifka MK. Optimization of peptide-based ELISA for serological diagnostics: a retrospective study of human monkeypox infection. Vector Borne Zoonotic Dis. 2012;12(5):400–9.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Jezek Z, Nakano JH, Arita I, Mutombo M, Szczeniowski M, Dunn C. Serological survey for human monkeypox infections in a selected population in Zaire. J Trop Med Hyg. 1987;90(1):31–8.

    CAS  PubMed  Google Scholar 

  103. Dubois ME, Slifka MK. Retrospective analysis of monkeypox infection. Emerg Infect Dis. 2008;14(4):592–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Kramski M, Drozd A, Lichtfuss GF, Dabrowski PW, Ellerbrok H. Rapid detection of anti-Vaccinia virus neutralizing antibodies. Virol J. 2011;8:139.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Esposito JJ, Obijeski JF, Nakano JH. The virion and soluble antigen proteins of variola, monkeypox, and vaccinia viruses. J Med Virol. 1977;1(2):95–110.

    Article  CAS  PubMed  Google Scholar 

  106. Hutson CL, Gallardo-Romero N, Carroll DS, Clemmons C, Salzer JS, Nagy T, Hughes CM, Olson VA, Karem KL, Damon IK. Transmissibility of the monkeypox virus clades via respiratory transmission: investigation using the prairie dog-monkeypox virus challenge system. PLoS ONE. 2013;8(2):e55488.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Fernández de Marco Mdel M, Alejo A, Hudson P, Damon IK, Alcami A: The highly virulent variola and monkeypox viruses express secreted inhibitors of type I interferon. Faseb J. 2010;24(5):1479–1488.

  108. Baxby D. The surface antigens of orthopoxviruses detected by cross-neutralization tests on cross-absorbed antisera. J Gen Virol. 1982;58(Pt 2):251–62.

    Article  CAS  PubMed  Google Scholar 

  109. McCollum AM, Damon IK. Human monkeypox. Clin Infect Dis. 2014;58(2):260–7.

    Article  PubMed  Google Scholar 

  110. Dabrowski PW, Radonić A, Kurth A, Nitsche A. Genome-wide comparison of cowpox viruses reveals a new clade related to Variola virus. PLoS ONE. 2013;8(12):e79953.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Huggett JF, French D, O’Sullivan DM, Moran-Gilad J, Zumla A. Monkeypox: another test for PCR. Euro Surveill. 2022;27(32):2200497.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. WHO | Laboratory testing for the monkeypox virus: interim guidance, 23 May 2022. available from: https://www.who.int/publications/i/item/WHO-MPX-laboratory-2022.1. Accessed 10 Sept 2024.

  113. Iizuka I, Saijo M, Shiota T, Ami Y, Suzaki Y, Nagata N, Hasegawa H, Sakai K, Fukushi S, Mizutani T, et al. Loop-mediated isothermal amplification-based diagnostic assay for monkeypox virus infections. J Med Virol. 2009;81(6):1102–8.

    Article  CAS  PubMed  Google Scholar 

  114. Piepenburg O, Williams CH, Stemple DL, Armes NA. DNA detection using recombination proteins. PLoS Biol. 2006;4(7):e204.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Glökler J, Lim TS, Ida J, Frohme M. Isothermal amplifications - a comprehensive review on current methods. Crit Rev Biochem Mol Biol. 2021;56(6):543–86.

    Article  PubMed  Google Scholar 

  116. Zhao F, Wang P, Wang H, Liu S, Sohail M, Zhang X, Li B, Huang H: CRISPR/Cas12a-mediated ultrasensitive and on-site monkeypox viral testing. medRxiv. 2022:2022.2010.2010.22280931.

  117. Amer F, Khalil HES, Elahmady M, ElBadawy NE, Zahran WA, Abdelnasser M, Rodríguez-Morales AJ, Wegdan AA, Tash RME. Mpox: Risks and approaches to prevention. J Infect Public Health. 2023;16(6):901–10.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Al-Qerem W, Alassi A, Jarab A, Al Bawab AQ, Hammad A, Alasmari F, Alazab B, Abu Husein D, Al Momani N, Eberhardt J. Examining Influenza Vaccination Patterns Among Young Adults with Asthma: Insights into Knowledge, Attitudes, and Practices. Patient Prefer Adherence. 2023;17:2899–913.

    Article  PubMed  PubMed Central  Google Scholar 

  119. CDC | Vaccine-Preventable Diseases; available from: https://www.cdc.gov/global-immunization/diseases/index.html. Accessed 14 Sept 2024.

  120. CDC | Mpox-Vaccination; available from: https://www.cdc.gov/poxvirus/mpox/interim-considerations/overview.html#:~:text=Resources-,Vaccines,to%20orthopoxvirus%20infections%2C%20including%20mpox. Accessed 14 Sept 2024.

  121. WHO | Vaccine-Preventable Diseases (including pipeline vaccines); available from: https://www.who.int/teams/immunization-vaccines-and-biologicals/diseases. Accessed 14 Sept 2024.

  122. Hajjo R, Sabbah DA, Bardaweel SK, Abusara OH. Epidemiology Analysis of COVID-19 Data and Associated Effects of Pandemic Containment Procedures, Viral Variants, and Vaccines: Lessons Learned From Middle Eastern Jordan’s Experience. Advances in Public Health. 2024;2024(1):2977631.

    Article  Google Scholar 

  123. Ghazy RM, Elrewany E, Gebreal A, ElMakhzangy R, Fadl N, Elbanna EH, Tolba MM, Hammad EM, Youssef N, Abosheaishaa H, et al. Systematic Review on the Efficacy, Effectiveness, Safety, and Immunogenicity of Monkeypox Vaccine. Vaccines (Basel). 2023;11(11):1708.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Volz A, Sutter G. Modified Vaccinia Virus Ankara: History, Value in Basic Research, and Current Perspectives for Vaccine Development. Adv Virus Res. 2017;97:187–243.

    Article  CAS  PubMed  Google Scholar 

  125. Geha RM, Chen K, Wouters J, Ooms F, Shih JC. Analysis of conserved active site residues in monoamine oxidase A and B and their three-dimensional molecular modeling. J Biol Chem. 2002;277(19):17209–16.

    Article  CAS  PubMed  Google Scholar 

  126. Li M, Binda C, Mattevi A, Edmondson DE. Functional role of the “aromatic cage” in human monoamine oxidase B: structures and catalytic properties of Tyr435 mutant proteins. Biochemistry. 2006;45(15):4775–84.

    Article  CAS  PubMed  Google Scholar 

  127. Malone SM, Mitra AK, Onumah NA, Brown A, Jones LM, Tresvant D, Brown CS, Onyia AU, Iseguede FO: Safety and Efficacy of Post-Eradication Smallpox Vaccine as an Mpox Vaccine: A Systematic Review with Meta-Analysis. Int J Environ Res Public Health. 2023;20(4).

  128. Ladhani SN, Dowell AC, Jones S, Hicks B, Rowe C, Begum J, Wailblinger D, Wright J, Owens S, Pickering A, et al. Early evaluation of the safety, reactogenicity, and immune response after a single dose of modified vaccinia Ankara-Bavaria Nordic vaccine against mpox in children: a national outbreak response. Lancet Infect Dis. 2023;23(9):1042–50.

    Article  CAS  PubMed  Google Scholar 

  129. van der Boom M, van Hunsel F. Adverse reactions following MPox (monkeypox) vaccination: An overview from the Dutch and global adverse event reporting systems. Br J Clin Pharmacol. 2023;89(11):3302–3310.

    Article  PubMed  Google Scholar 

  130. Tomita N, Terada-Hirashima J, Uemura Y, Shimizu Y, Iwasaki H, Yano R, Suzuki T, Saito S, Okumura N, Sugiura W, et al. An open-label, non-randomized study investigating the safety and efficacy of smallpox vaccine, LC16, as post-exposure prophylaxis for mpox. Hum Vaccin Immunother. 2023;19(2):2242219.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Lin YC, Wen TH, Shih WL, Vermund SH, Fang CT. Impact of vaccination and high-risk group awareness on the mpox epidemic in the United States, 2022–2023: a modelling study. EClinicalMedicine. 2024;68:102407.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Bourner J, Redji Mbrenga FD, Malaka CN, Dunning J, Rojek A, Fandema E, Horby P, Boum Y 2nd, Nakouné E, Olliaro P. Expanded Access Programme for the use of tecovirimat for the treatment of monkeypox infection: A study protocol for an Expanded Access Programme. PLoS ONE. 2024;19(5): e0278957.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Matias WR, Koshy JM, Nagami EH, Kovac V, Moeng LR, Shenoy ES, Hooper DC, Madoff LC, Barshak MB, Johnson JA, et al. Tecovirimat for the Treatment of Human Monkeypox: An Initial Series From Massachusetts, United States.Open forum infectious diseases. 2022;9(8):ofac377.

  134. Desai AN, Thompson GR 3rd, Neumeister SM, Arutyunova AM, Trigg K, Cohen SH. Compassionate Use of Tecovirimat for the Treatment of Monkeypox Infection. JAMA. 2022;328(13):1348–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. O’Laughlin K, Tobolowsky FA, Elmor R, Overton R, O’Connor SM, Damon IK, Petersen BW, Rao AK, Chatham-Stephens K, Yu P, et al. Clinical Use of Tecovirimat (Tpoxx) for Treatment of Monkeypox Under an Investigational New Drug Protocol - United States, May-August 2022. MMWR Morb Mortal Wkly Rep. 2022;71(37):1190–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Wu EL, Osborn RL, Bertram CM, Moore WJ, Galvin SR, Bolon MK, Masters MC, Krueger KM. Tecovirimat Use in Ambulatory and Hospitalized Patients With Monkeypox Virus Infection. Sex Transm Dis. 2023;50(3):172–4.

    Article  CAS  PubMed  Google Scholar 

  137. Chittick G, Morrison M, Brundage T, Nichols WG. Short-term clinical safety profile of brincidofovir: A favorable benefit–risk proposition in the treatment of smallpox. Antiviral Res. 2017;143:269–77.

    Article  CAS  PubMed  Google Scholar 

  138. Chan-Tack K, Harrington P, Bensman T, Choi S-Y, Donaldson E, O'Rear J, McMillan D, Myers L, Seaton M, Ghantous H, et al. Benefit-risk assessment for brincidofovir for the treatment of smallpox: U.S. Food and Drug Administration's Evaluation. Antiviral Res. 2021;195:105182.

  139. Alvarez-Cardona JJ, Whited LK, Chemaly RF. Brincidofovir: understanding its unique profile and potential role against adenovirus and other viral infections. Future Microbiol. 2020;15:389–400.

    Article  CAS  PubMed  Google Scholar 

  140. Chamberlain JM, Sortino K, Sethna P, Bae A, Lanier R, Bambara RA, Dewhurst S. Cidofovir Diphosphate Inhibits Adenovirus 5 DNA Polymerase via both Nonobligate Chain Termination and Direct Inhibition, and Polymerase Mutations Confer Cidofovir Resistance on Intact Virus. Antimicrob Agents Chemother. 2019;63(1):e01925-e1918.

    Article  CAS  PubMed  Google Scholar 

  141. Baker RO, Bray M, Huggins JW. Potential antiviral therapeutics for smallpox, monkeypox and other orthopoxvirus infections. Antiviral Res. 2003;57(1):13–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Smee DF. Progress in the Discovery of Compounds Inhibiting Orthopoxviruses in Animal Models. Antivir Chem Chemother. 2008;19(3):115–24.

    Article  CAS  PubMed  Google Scholar 

  143. Rice AD, Adams MM, Wallace G, Burrage AM, Lindsey SF, Smith AJ, Swetnam D, Manning BR, Gray SA, Lampert B, et al. Efficacy of CMX001 as a Post Exposure Antiviral in New Zealand White Rabbits Infected with Rabbitpox Virus, a Model for Orthopoxvirus Infections of Humans. Viruses. 2011;3(1):47–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Parker S, Chen NG, Foster S, Hartzler H, Hembrador E, Hruby D, Jordan R, Lanier R, Painter G, Painter W, et al. Evaluation of disease and viral biomarkers as triggers for therapeutic intervention in respiratory mousepox – An animal model of smallpox. Antiviral Res. 2012;94(1):44–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. ClinicalTrials.gov. Identifier: NCT04706923. A Phase 2a Study of IV BCV in Subjects With Adenovirus Infection (ATHENA); available from: https://www.clinicaltrials.gov/study/NCT04706923. Accessed 31 Oct 2024.

  146. ClinicalTrials.gov. Identifier: NCT01143181. Study to Assess Brincidofovir Treatment of Serious Diseases or Conditions Caused by Double-stranded DNA Viruses; available from: https://clinicaltrials.gov/study/NCT01143181. Accessed 31 Oct 2024.

  147. Guzman AK, Schairer DO, Garelik JL, Cohen SR. Safety and efficacy of topical cantharidin for the treatment of pediatric molluscum contagiosum: a prospective, randomized, double-blind, placebo-controlled pilot trial. Int J Dermatol. 2018;57(8):1001–6.

    Article  CAS  PubMed  Google Scholar 

  148. Cinatl J, Bechtel M, Reus P, Ott M, Rothweiler F, Michaelis M, Ciesek S, Bojkova D. Trifluridine for treatment of mpox infection in drug combinations in ophthalmic cell models. J Med Virol. 2024;96(1):e29354.

    Article  CAS  PubMed  Google Scholar 

  149. Perzia B, Theotoka D, Li K, Moss E, Matesva M, Gill M, Kibe M, Chow J, Green S. Treatment of ocular-involving monkeypox virus with topical trifluridine and oral tecovirimat in the 2022 monkeypox virus outbreak. American journal of ophthalmology case reports. 2023;29:101779.

    Article  PubMed  Google Scholar 

  150. Kaufman AR, Chodosh J, Pineda R II. Monkeypox Virus and Ophthalmology—A Primer on the 2022 Monkeypox Outbreak and Monkeypox-Related Ophthalmic Disease. JAMA Ophthalmol. 2023;141(1):78–83.

    Article  PubMed  Google Scholar 

  151. Clarivate | Cortellis Drug Discovery Intelligence (CDDI); available from: https://clarivate.com/products/biopharma/research-development/pre-clinical-intelligence-analytics/. Accessed 21 Sept 2024.

  152. Ward BM, Riccio DA, Cartwright M, Maeda-Chubachi T. The Antiviral Effect of Berdazimer Sodium on Molluscum Contagiosum Virus Using a Novel In Vitro Methodology. Viruses. 2023;15(12):2360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Olatunji G, Kokori E, Kwape JM, Fawehinmi P, Stanley AC, Oluwatobiloba AM, Abraham IC, Shimelis KM, Olafimihan A, Ezeano C, et al. Berdazimer: A novel nitric oxide therapy in treating molluscum contagiosum. J Med Surg Public Health. 2024;2:100061.

    Article  Google Scholar 

  154. Remon J, Steuer CE, Ramalingam SS, Felip E: Osimertinib and other third-generation EGFR TKI in EGFR-mutant NSCLC patients. Ann Oncol. 2018;29(suppl_1):i20-i27.

  155. Capriotti K, Stewart K, Pelletier J, Capriotti J. Molluscum Contagiosum Viral Infection Treated With a Dilute Povidone-Iodine/Dimethylsulfoxide Preparation. Dermatol Ther. 2016;6(1):101–3.

    Article  Google Scholar 

  156. Tarrand Jeffrey J, LaSala PR, Han X-Y, Rolston Kenneth V, Kontoyiannis Dimitrios P. Dimethyl Sulfoxide Enhances Effectiveness of Skin Antiseptics and Reduces Contamination Rates of Blood Cultures. J Clin Microbiol. 2020;50(5):1552–7.

    Article  Google Scholar 

  157. NCIthesaurus| East Indian Sandalwood Oil Cream (Code C150380); available from: https://ncit.nci.nih.gov/ncitbrowser/ConceptReport.jsp?dictionary=NCI_Thesaurus&ns=ncit&code=C150380. Accessed 6 Dec 2024.

  158. AdisInsight| Santalum album cream - Santalis Healthcare; available from: https://adisinsight.springer.com/drugs/800033062. Accessed 6 Dec 2024.

  159. Zuiani A, Dulberger CL, De Silva NS, Marquette M, Lu YJ, Palowitch GM, Dokic A, Sanchez-Velazquez R, Schlatterer K, Sarkar S, et al. A multivalent mRNA monkeypox virus vaccine (BNT166) protects mice and macaques from orthopoxvirus disease. Cell. 2024;187(6):1363-1373.e1312.

    Article  CAS  PubMed  Google Scholar 

  160. ClinicalTrial.gov| A Clinical Study Investigating the Safety and Immune Responses After Immunization With Investigational Monkeypox Vaccines; available from: https://clinicaltrials.gov/study/NCT05988203. Accessed 6 Dec 2024.

  161. PR-Newswire | Ascletis Announces FDA Clearance of Oral RdRp Inhibitor ASC10 to Conduct a Randomized, Placebo Controlled Phase Ib Study in Mild-to-Moderate COVID-19 Patients; available from: https://www.prnewswire.com/news-releases/ascletis-announces-fda-clearance-of-oral-rdrp-inhibitor-asc10-to-conduct-a-randomized-placebo-controlled-phase-ib-study-in-mild-to-moderate-covid-19-patients-301598784.html. Accessed 15 Sept 2024.

  162. PR-Newswire | Ascletis Announces Positive Phase I Clinical Results of Oral RdRp Inhibitor ASC10 for COVID-19; available from: https://www.prnewswire.com/news-releases/ascletis-announces-positive-phase-i-clinical-results-of-oral-rdrp-inhibitor-asc10-for-covid-19-301700402.html. Accessed 15 Sept 2024.

  163. Chakraborty A, Diwan A, Tatake J. Prospect of nanomaterials as antimicrobial and antiviral regimen. AIMS microbiology. 2023;9(3):444–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. ContagionLive | A Single Therapy Against Multiple Respiratory Virus Infections?; available from: https://www.contagionlive.com/view/a-single-therapy-against-multiple-respiratory-virus-infections-. Accessed 15 Sept 2024.

  165. Barnette KG, Gordon MS, Rodriguez D, Bird TG, Skolnick A, Schnaus M, Skarda PK, Lobo S, Sprinz E, Arabadzhiev G, et al. Oral Sabizabulin for High-Risk, Hospitalized Adults with Covid-19: Interim Analysis. NEJM evidence. 2022;1(9):EVIDoa2200145.

  166. Markowski MC, Tutrone R, Pieczonka C, Barnette KG, Getzenberg RH, Rodriguez D, Steiner MS, Saltzstein DR, Eisenberger MA, Antonarakis ES. A Phase Ib/II Study of Sabizabulin, a Novel Oral Cytoskeleton Disruptor, in Men with Metastatic Castration-resistant Prostate Cancer with Progression on an Androgen Receptor-targeting Agent. Clinical cancer research : an official journal of the American Association for Cancer Research. 2022;28(13):2789–95.

    Article  CAS  PubMed  Google Scholar 

  167. Labiotech | Emergex develops vaccine for smallpox and monkeypox; available from: https://www.labiotech.eu/trends-news/emergex-creates-vaccine-smallpox-monkeypox/#:~:text=Emergex%20Vaccines%20Holding%20Limited%20has,early%20%E2%80%9Ceclipse%20phase%E2%80%9D%20antigens. Accessed 19 Oct 2024.

  168. Tonix Pharmaceuticals. Tonix Pharmaceuticals Provides Update on the Development of Its Single Dose Live Attenuated Virus Vaccine Candidate for Mpox, TNX-801, as WHO Declares Mpox Outbreak a Global Health Emergency; available from: https://ir.tonixpharma.com/news-events/press-releases/detail/1505/tonix-pharmaceuticals-provides-update-on-the-development-of. Accessed 19 Oct 2024.

  169. NCBI | NCBI Virus; available from: https://www.ncbi.nlm.nih.gov/labs/virus/vssi/#/. Accessed 21 Sept 2024.

  170. Yue Y, Jiang M, Zhang X, Xu J, Ye H, Zhang F, Li Z, Li Y. Mpox-AISM: AI-mediated super monitoring for mpox and like-mpox. iScience. 2024;27(5):109766.

  171. Cheng K, He Y, Li C, Xie R, Lu Y, Gu S, Wu H. Talk with ChatGPT About the Outbreak of Mpox in 2022: Reflections and Suggestions from AI Dimensions. Ann Biomed Eng. 2023;51(5):870–4.

    Article  PubMed  PubMed Central  Google Scholar 

  172. WHO| Emergencies; available from: https://www.who.int/europe/emergencies/resources. Accessed 7 Oct 2024.

  173. CDC| Information For Healthcare Professionals; available from: https://www.cdc.gov/poxvirus/mpox/clinicians/index.html. Accessed 7 Oct 2024.

Download references

Acknowledgements

R.H. and D.A.S. acknowledge funding from the Deanship of Scientific Research at Al-Zaytoonah University of Jordan (Grant Number 2023 - 2022/17/50).

Funding

Funding provided by the Deanship of Scientific Research at Al-Zaytoonah University of Jordan (Grant Number 2023 - 2022/17/50).

Author information

Authors and Affiliations

Authors

Contributions

Idea and conceptualization, network and pathway analysis, genetic epidemiology analysis, writing first draft, editing manuscript and reviewing literature (R.H.). Writing first draft, editing, collecting data and reviewing previous literature (O.A.S). Writing first draft, editing, collecting data and reviewing literature (D.A.S.). Idea, writing first draft, editing, collecting data and reviewing previous literature (S.K.B).

Corresponding author

Correspondence to Rima Hajjo.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hajjo, R., Abusara, O.H., Sabbah, D.A. et al. Advancing the understanding and management of Mpox: insights into epidemiology, disease pathways, prevention, and therapeutic strategies. BMC Infect Dis 25, 529 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12879-025-10899-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12879-025-10899-2

Keywords