Skip to main content

Current status of hand-foot-and-mouth disease

Abstract

Hand-foot-and-mouth disease (HFMD) is a viral illness commonly seen in young children under 5 years of age, characterized by typical manifestations such as oral herpes and rashes on the hands and feet. These symptoms typically resolve spontaneously within a few days without complications. Over the past two decades, our understanding of HFMD has greatly improved and it has received significant attention. A variety of research studies, including epidemiological, animal, and in vitro studies, suggest that the disease may be associated with potentially fatal neurological complications. These findings reveal clinical, epidemiological, pathological, and etiological characteristics that are quite different from initial understandings of the illness. It is important to note that HFMD has been linked to severe cardiopulmonary complications, as well as severe neurological sequelae that can be observed during follow-up. At present, there is no specific pharmaceutical intervention for HFMD. An inactivated Enterovirus A71 (EV-A71) vaccine that has been approved by the China Food and Drug Administration (CFDA) has been shown to provide a high level of protection against EV-A71-related HFMD. However, the simultaneous circulation of multiple pathogens and the evolution of the molecular epidemiology of infectious agents make interventions based solely on a single agent comparatively inadequate. Enteroviruses are highly contagious and have a predilection for the nervous system, particularly in child populations, which contributes to the ongoing outbreak. Given the substantial impact of HFMD around the world, this Review synthesizes the current knowledge of the virology, epidemiology, pathogenesis, therapy, sequelae, and vaccine development of HFMD to improve clinical practices and public health efforts.

Introduction

As early as 1957, the characteristic symptoms of fever, vesicular rash on hands and feet caused by Coxsackievirus (CV), primarily CVA16, was first reported in Toronto [1, 2]. In 1959, “hand-foot-and-mouth disease (HFMD)” was initially used to name a disease with essentially the same symptoms as described by Robinson et al. [3]. Over the past few decades, HFMD outbreaks caused by Enterovirus A71 (EV-A71), CVA16, CVA6 and Echoviruses (Echo) were reported frequently around the world [4]. EV-A71, which was first isolated from a child with meningitis in 1969, has also caused widespread outbreaks of HFMD throughout much of the Asia–Pacific region [5]. The disease was generally mild and lasted less than a week in most cases, characterized by fever, a blister-like rash on the hands and feet, and oral ulcers caused by ruptured blisters in the mouth [3]. However, quite a few patients experience fatal neurological or cardiopulmonary complications. Furthermore, recent follow-up studies have shown that severe neurological sequelae may occur in severely recovered patients (Fig. 1) [6,7,8]. Therefore, HFMD has become a significant concern for public health throughout the Asia–Pacific region and beyond. The discovery of tomato flu, a HFMD-like illness caused by enterovirus, in India has brought renewed attention to HFMD outbreaks [9]. This Review focuses on summarizing the current findings regarding HFMD in regards to virology, epidemiology, pathogenesis, and vaccine development in order to better inform clinical practice and public health initiatives.

Fig. 1
figure 1

Complications and sequelae of HFMD

Etiological characteristics of HFMD

HFMD is caused by Human enteroviruses (EVs) that are members of the Enterovirus genus of the Picornaviridae family [10]. EVs were initially classified into Poliovirus (PV), Echo, CV-A and B, and emerging EVs. Since 1999, EVs have been divided into four categories of Enterovirus A, B, C, and D, in the light of their molecular, biological, and genetic characteristics. Nowadays, over 100 EVs have been reported worldwide [10]. Table 1 lists various pathogens associated with HFMD outbreaks [11]. In the past, EV-A71 and CVA16 were the most frequently reported causes of HFMD prior to 2005. Currently, other EVs such as CVA6 and CVA10 are responsible for a significant proportion of HFMD cases and outbreaks [12, 13]. Although CVB1-5 associated with HFMD had also been mentioned in several reports, the impact of these pathogens on HFMD is still expanding [14, 15].

Table 1 EVs associated with HFMD

The viral particle of EVs is symmetrical icosahedron composed of 60 subunits of coat protein and a single-stranded RNA genome (7.5 kb) of positive polarity (Fig. 2) [16]. The open reading frame (ORF) of the viral genome encodes 2194 amino acids (Fig. 2), and the 3′ untranslated region UTR (3'UTR) is followed by a poly-A tail of variable length. The protein encoded by the viral genome mainly include three regions: P1, P2, and P3, of which P1 encodes four structural proteins, VP1-VP4, and the P2 and P3 encode seven non-structural proteins, 2A-2C and 3A-3D, respectively [17, 18]. VP1-VP4 are further involved in virion capsid assembly [17]. Although VP1, VP2 and VP3 are arranged on the outer side of the capsid, VP1 is the main antigen-binding site [19]. Thus, VP1 is a suitable candidate for major serotyping and vaccine development and has been widely used as a target gene for EVs molecular research [20]. Moreover, Physico-chemical characteristics of EVs include resistance to organic solvents such as ether and chloroform and low temperature conditions, and sensitivity to high temperature, chlorinated disinfectants, formaldehyde and ultraviolet etc. [10].

Fig. 2
figure 2

The life cycle of Enterovirus. Enterovirus (EVs) enter the host cells by binding to receptors or by exosome-mediated endocytosis and release positive-strand RNA. The RNA undergoes transcription and translation after being covalently linked to the viral protein VPg (3B). The translated polyprotein is hydrolyzed by various proteases into 10 separate major proteins, including VP0, VP1, VP3, 2A-C, 3A-D, where VP0 is subsequently hydrolyzed to VP2 and VP4. VP1-4 are assigned to participate in the assembly of viral protein coats, while 2A-C, 3A-D are directed to participate in the replication of viral genetic material. Finally, the viral RNA and coat are assembled and processed into mature viruses, which are then co-packaged with host organelle decomposers in vesicles and secreted out of the cell, or directly released by exocytosis

Epidemiological characteristics

Epidemic process and influencing factors

Clinical features

The criteria for diagnosing of HFMD, which are widely accepted at present, primarily rely on the patient’s epidemiological history, symptoms additional tests to determine the cause or presence of the disease [21]. This includes examination of the patient's age, the timing of onset, gathering place, and if they had direct or indirect contact with HFMD infections before the onset of the disease [21]. The incubation period of HFMD is mostly 2–10 days, with an average of 3–5 days. The progression of HFMD be divided into 5 stages (rash, neurological dysfunction, early stage of cardiopulmonary failure, cardiopulmonary failure, recovery), and most cases generally only experience the first stage and recover within a week [22]. Clinically, most cases have fever accompanied by rash on hands, feet, mouth, and buttocks [21]. The prevention in patients with severe HFMD depends on the timely and accurate identification of danger signs in the disease progression [21]. The following 7 indicators are considered as risk factors of HFMD severity: (1) high fever; (2) nervous system involvement; (3) abnormal respiratory rate and rhythm; (4) circulatory dysfunction; (5) increased white blood cell count; (6) increased blood glucose; (7) increased blood lactate [21, 23]. In some cases of HFMD, the rash is atypical such as a single site or a maculopapular rash only. Most cases usually need to be differentiated from papular urticaria, chickenpox, herpes zoster, rubella, and herpes simplex caused by other diseases [21]. In addition, neurogenic pulmonary edema (PE) should be distinguished from pneumonia. Clinical samples (pharyngeal swabs, stool or anal swabs, blood, blister fluid, cerebrospinal fluid, etc.) are tested through RT-PCR, virus isolation, neutralizing antibody testing [21]. Subsequently, clinicians diagnosed the suspected patient as a confirmed case of HFMD based on epidemiological history, clinical manifestations, and laboratory nucleic test [24].

Source of infection

Human is usually considered to be the only reservoir of human EVs, and both cases and asymptomatic infections are the sources of HFMD infection. The virus can be detected in the pharynx and feces of infected individuals in the days before the onset of illness, and is usually most contagious within a week after the onset of symptoms. Therefore, the presence of asymptomatic infections and those in the incubation period may complicate efforts to prevent and control HFMD.

Routes of transmission

Currently, the fecal–oral transmission and contact are considered as the primary transmission routes of HFMD. The potential transmission routes of aerosols and respiratory tract have been proposed based on some animal studies [25]. Further research is required to fully understand and confirm the transmission routes of both aerosol and droplet in human population.

Herd susceptibility

As a common childhood infectious disease, HFMD primarily occurs in children under 5 years old [26], although HFMD has also been reported in adults [27]. Children are highly susceptible to the EVs due to immature immune system and clustering at the pre-kindergarten stage [28]. China has implemented kindergarten closures to block the transmission of coronavirus disease (COVID-19), indirectly reducing the incidence of HFMD and preventing HFMD outbreaks [29, 30]. In addition to reducing the clustering of susceptible populations and enhancing individual protection, “herd” immunity through vaccination is more effective in reducing population susceptibility. The urban area with high EV-A71 transmission in China initiated vaccination with inactivated EV-A71 vaccine, a dramatic decline in EV-A71-associated HFMD incidence was observed [31]. Patients, both dominant and recessive infections caused by EVs, can acquire specific immunity, and the neutralizing antibodies can be retained in the body for a long time. EVs can stimulate stronger immune response, but there is almost no cross-immunity between different serotypes. Consequently, multivalent vaccines are urgently needed to further improve herd immunity.

Spectrum of infection

HFMD is always considered as a type of self-limiting infectious disease, and most patients with mild symptoms recover within 1 ~ 2 weeks. Large-scale observational studies showed that There are 5 different outcomes of HFMD: asymptomatic (12.7%), mild (86.2%), severe and critical (1.1%), death (0.03%) [24], [32].

Natural factors

Both high and low temperatures were associated with the incidence of HFMD [33]. For example, CVA6 outbreaks usually occur in winter [34]. Precipitation and humidity could provide the necessary water environment and aerosols for virus survival, and protect the virus from harmful factors such as temperature, salinity, and pH [35]. The intensity of UV exposure time also affected the incidence of HFMD [36]. Also, the terrain dominated by mountains or hills with lower atmospheric pressure affects the incidence of HFMD [37]. Recently, a new attention has been attracted to the impact of air pollution on HFMD epidemic. Yu et al. found that exposure to environmental particulate matter increases the risk of children developing HFMD. They believed that these particulates may facilitate virus transmission through airborne infections and that high wind speeds further contribute to the spread of virus-carrying particles [28]. Besides, ozone might affect infectious diseases by inhibiting the ability of virus to exist in the external environment [28].

Socioeconomic factors

Socioeconomic factors are also closely related to the epidemic of HFMD. The incidence of HFMD in urban residents, transportation hub cities, and economically developed areas compared to rural area, this is due to the higher population density and mobility in these areas [26, 38]. Health regulations and large-scale vaccination in educational settings promulgated by the state or government at all levels significantly reduced the incidence of HFMD [28]. The lack of medical insurance coverage and ethnic minorities are all risk factors for HFMD [26]. Rural residents and poverty are both risk factors for HFMD severity, which may be caused by poor sanitation, lower educational attainment, and lower economic status [26]. Furthermore, factors such as being raised at home, having a larger family size, and poor hand hygiene are associated with a higher risk of HFMD transmission [39, 40]. Short interval from onset to hospitalization, hospitalization in a high-level hospital, and treatment by more experienced doctor are protective factors for HFMD severity [41]. Lack of breastfeeding in children with lower immune status may lead to HFMD severity [42, 43]. Extended gatherings of children in schools or daycare centers can facilitate the transmission of HFMD, while taking appropriate breaks during vacation time can serve as a protective measure against it [44].

The four main EV serotypes causing HFMD outbreaks

HFMD of outbreaks caused by EV-A71

The EV-A71 strain was first isolated in California in 1969 [5]. During 1970–1990, HFMD outbreaks caused by EV-A71 occurred frequently in the United States [45,46,47]. In the European, including Sweden [48], Bulgaria [49], Hungary [50], and the Netherlands [51], outbreaks of HFMD related to EV-A71 have been monitored. Japan [52], Brazil [53] and Australia have also reported a large number of cases of aseptic meningitis and brainstem encephalitis associated with EV-A71. At the end of the twentieth century, EV-A71 activity increased dramatically throughout the Western Pacific region. In 1997, a large outbreak of HFMD caused by EV-A71 strain in Malaysia resulted in 41 fatalities [54]. Next year, Taiwan (China) reported 100,000 cases of HFMD mainly caused by EV-A71, including 400 severe cases and 78 deaths [55]. During the period from 2008 to 2014, a total of 10,717,283 cases (3046 deaths) were reported in China, and the fatality rate was 0.03%. Among survivors, the incidence increased from 37.6/100,000 (2008) to 139.6/100,000 (2013) and had a peak in 2012 at 166.8/100,000. In 2011–2012, a large-scale EV-A71 outbreak in Vietnam resulted in more than 200,000 hospitalizations and 207 deaths [56]. In 2012, EV-A71 infection killed at least 54 children with severe encephalitis in Cambodia (26,690,000). In addition, Russia [57], South Korea [58], Singapore [59], Thailand [60, 61] and Philippines [62] have also experienced large-scale EV-A71 outbreaks. Recently, European countries such as Denmark [63], France [64], Germany [65], Spain [66] and Poland [67] also reported sporadic cases (Fig. 3).

Fig. 3
figure 3

Distribution of patients with HFMD in the world. A EV-A71; B CVA16; C CVA6; D CVA10. Areas marked in orange indicate that EV-A71/CVA16/CVA6/CVA10 epidemic have been reported

HFMD of outbreaks caused by CVA16

CVA16 was the main pathogen of HFMD outbreaks in England in 1959 and 1994 [3, 68]. There were also CVA16 outbreaks in the United States in 1964 and 1968 [69, 70]. CVA16 infection was also responsible for the 1991 outbreak of HFMD in Sydney, Australia [71]. Subsequently, the Asian-Pacific region includes China [72, 73], Japan [74], India [75, 76], Taiwan (China) [77], Vietnam [78], Singapore [79, 80], and Spain in Europe [81] reported CVA16 outbreaks. Currently, CVA16 pathogens are frequently detected together with CVA6 and CVA10 [43, 82,83,84].

HFMD outbreaks caused by CVA6

In recent years, the pathogenic spectrum of HFMD has changed with inoculation of EV-A71 vaccines, especially in China. From 2016 to 2018, the proportion of EV-A71 and CVA16 positive was 8.9%, 5.2%, respectively, while the proportion of other EVs was 60.6% among 3559 HFMD cases in Hangzhou, China [85]. Since an outbreak of HFMD caused by CVA6 in Finland in 2008, CVA6 is responsible for a series of HFMD outbreaks in Europe, Northern America, and Asia [86]. In recent years, HFMD outbreaks caused by CVA6 have occurred in the United States [87, 88], Spain [81], Hungary [89], France [84] and the United Kingdom (Fig. 3) [90]. EVs were detected in 2228 HFMD patients in Vietnam from 2008 to 2017, and CVA6 accounted for 28.4%, only second to EV-A71 (31.7%). However, the large-scale HFMD outbreak in Thailand in 2012 showed that in 672 HFMD cases, 221 (32.9%) were caused by CVA6 [91]. In 2011, the National Epidemiological Surveillance System of Infectious Diseases of Japan reported an increase rate of CVA6 detection in HFMD cases [92]. In the massive HFMD epidemic that occurred in Japan in 2017, CVA6 was the primary pathogen responsible for the illness of 6,173 patients [93]. In addition, Singapore [94], New Zealand [95] and Malaysia [96] have also reported HFMD CVA6 is the dominant strain of HFMD outbreaks. Since 2013, CVA6-associated HFMD has been on the rise in parts of China [97,98,99,100]. Unlike HFMD caused by other EVs serotypes, CVA6-associated HFMD presents a more severe and extensive rash, and is also characterized by a higher incidence in adults, winter onset, and a tendency to shed arm after recovery [34, 101, 102].

HFMD of outbreaks caused by CVA10

The prototype strain of CVA10, Kowalik (GenBank ID: AY421767), was isolated in the United States in 1950 [103]. In May 1961, the CVA10 strain was also isolated in 40 children with HFMD reported in New Zealand [104]. The first detailed outbreak of CVA10 occurred in Japan between July 1981 and January 1982. Thirty seven clinical HFMD cases were examined for virology and serology, and CVA10 was detected in 18 cases [105]. Subsequently, Asia, Europe, Africa, and Oceania successively reported HFMD associated with CVA10 co-transmitted with CVA6 (Fig. 3). In 2008, clinical specimens were obtained from 317 HFMD cases in Finland, including adults and children, and the proportion of CVA10 (28%) was only second to CVA6 (71%) [86]. The HFMD epidemic surveillance in Singapore in 2008 showed that the detection rate of CVA10 (11.8%) ranked third, followed by CVA6 (23.5%) and EV-A71 (21.6%) [94]. A French sentinel surveillance data study conducted in 2010 reported that CVA10 (39.9%) was the leading serotype responsible for HFMD [106]. In Asia, CVA10 was also the most common pathogen in HFMD cases monitored in Korea in 2008 [107]. In a prospective cohort study in 2016, a higher proportion of CVA10 was detected in HFMD cases [43]. There are different levels of CVA10 detection rates in HFMD patients across China. CVA10 (25%), CVA6 (29.8%), and CVA16 (32.5%) were the most common serotypes [108] in HFMD patients in Guangdong Province in 2018. In Xiamen, from 2009 to 2015, the proportion of CVA10 in cases of HFMD was not particularly high (1.08–7.09%). However, the detection rate of CVA10 in severe HFMD cases was significantly higher than in previous years [109]. From 2016 to 2020, a total of 9952 sporadic HFMD cases in Shanghai were collected and CVA10 was the fourth major epidemic pathogens, with a total positive rate of 2.78% [110].

Genetic evolution of EVs

EV-A71, CVA16, CVA6 and CVA10 are the 4 major EVs that cause HFMD worldwide. There are no standardized criteria for the classification of subtype, and the different studies on the prevalent types of HFMD use distinct system of sub‐type classification [111]. Bayesian phylogenetic methods with an integrated molecular clock were introduced a decade ago and provided unprecedented opportunities for phylogenetic analysis. In the Review, a difference of at least 15% in the entire VP1 nucleotide sequences is used to distinguish genotypes (Fig. 4) [112]. The sequences were used to identify the serotypes/sub-genotypes using the online Enterovirus Genotyping Tool (http://www.rivm.nl/mpf/enterovirus/typingtool) or a BLAST search. In the case of CVA6 and CVA10, we consult other studies to ensure that the selected strains are representative [110, 113]. The genetic evolution of EV-A71 virus can be divided into seven genotypes (A-G), with genotypes B and C further divided into sub-genotypes B0-B5 and C1-C5, respectively [112]. Genotype A includes the prototype strain (BrCr) isolated in 1969 [5]. C4 and C1 sub-genotypes have developed into the most predominant strain and sub-genotypes C4 circulate mainly in eastern and southeast Asia, whereas C1 are prevalent in Europe [64, 114]. D-G genotypes are relatively rare strains and have been identified in India [115], Africa [116] and Madagascar [117]. There are also several strains that can’t be typed in the online enterovirus Genotyping Tool (defined by some scholars C0: AF135934.1, H: ON646273.1). CVA16 is divided into 2 genogroups A and B with genogroup B being further divided into B1 and B2. Sub-genotype B1 can be further divided into clusters B1a, B1b, and B1c. B1a and B1b can be found in China, Malaysia, Thailand, Australia, Vietnam, and France, Japan et al., which indicate that they co-evolve and co-circulate all over the world [118, 119]. Recently, new genogroups (C and D named by some scholars) have been reported in Peru, France, and China [120,121,122]. Our results revealed that CVA6 strains could be divided into 6 genotypes designated as A to F, and D genotypes could be further subdivided into D1-3 sub-genotypes. In recent years, the D genotype, particularly D3 sub-genotype, has become the dominant sub-genotype circulating in Southeast Asia and Europe [20, 123]. CVA10 is assigned into 7 genogroups, including genogroup A to genogroup G. Genogroup A is the prototype Kowalik strain isolated in 1950 in the United States [124]. Genogroup B, mainly consisted of CVA10 in China, is assigned to genogroup G. Genogroup C and D include isolates from Russia, Viet Nam, France, America, as well as the latest isolates from Mainland China, which is the predominant circulating strain worldwide [110]. Genogroup E and F mainly circulate in India [125] and Russia [126]. A study on the prevalence of HFMD-associated EVs in China found that more than 98% of EV-A71 sequences belonged to the C4 sub-genotype, with the EV-A71-C4.1 strain having the largest proportion, the longest epidemic period, and the widest geographical distribution. The most predominant strain of CVA16 was CVA16-B1.1, which was widely found in East, Southern, and Northern China. Approximately 95.6% of CVA6 strains belonged to the D genotype and were mainly prevalent in the Eastern, Northern, and Southern regions of China. Most of the CVA10 strains in China belonged to the C sub-genotype and were mainly found in Eastern China [20]. Furthermore, recombination events between other EVs and increased detection rates of these EVs in HFMD samples have been a significant factor in recent HFMD outbreaks. A study of the genome sequence of a novel CVB2 (YN31V3) associated with HFMD found that YN31V3 was likely a recombinant, closely related to CVB2 strains and other EV-B strains [127]. The phylogenetic analysis of CVB3 sequences form the China national HFMD surveillance and global surveillance showed multiple recombination events were present among CVB3 strains circulating globally [128]. Taken together, for evolutionary pressure and frequent recombination, the pathogens of HFMD have evolved into a variety of EVs genotypes with specific temporal and spatial distributions, and further genomic analysis and continuous molecular epidemiological surveillance are helpful for disease control and prevention.

Fig. 4
figure 4

Phylogenetic analyses of the Enterovirus. Phylogenetic analyses of the EV-A71 (A), CVA16 (B), CVA6 (C) and CVA10 (D) circulating globally based on full length sequence of the VP1 gene worldwide available from GenBank were conducted in MEGA 7 using the neighbor-joining method. The bootstrap test was performed with 1000 replications. The evolutionary distances were written on the branch. We selected the representative VP1 sequences (EV-A71, n = 84; CVA16, n = 47; CVA6, n = 39; and CVA10, n = 56) from GenBank according to the country of origin, year of isolation and other information. All the strains are labelled using the following format: ‘accession number’/ ‘country of origin’/ ‘year of isolation’. All selected representative strains are marked with distinct colors according to different genotypes/sub-genotypes. The prototypes strains marked with yellow circles and red circles indicate the outgroups. The genotyping reference strains of different genotypes/sub-genotypes of CVA6 and CVA10 are marked as black triangles

Pathogenesis

The Viral receptors play a crucial role in the initial stage of infection. The first requirement for virus entry is to bind to the appropriate receptors on the host cells surface, triggering the next step of endocytosis. The availability of receptors often restricts viral infection and influence tissue and species specificity [129, 130]. Currently, most of receptors for EVs belong to the immunoglobulin superfamily (IgSF), which are type I transmembrane glycoproteins [131]. As summarized in Table 2, human scavenger receptor class B member 2 (hSCARB2) [132], P-selectin glycoprotein ligand-1 (PSGL-1) [133], Annexin II [134], Heparan sulfate [135] are identified to be the main receptors of EV-A71, and KREMEN1 was confirmed as a host entry receptor for CVA2, CVA3, CVA4, CVA5, CVA6, CVA7, CVA10, CVA14, CVA16 [136, 137]. EVs interact with host-encoded counterpart receptors and then undergo uncoating, pore formation, and release their genome into the cytosol [138]. EV-A71 binds to hSCARB2, and triggers a clathrin- and dynamin-dependent endocytosis to facilitate viral entry [139]. hSCARB2 and KREMEN1 bind to the canyons at the adaptor-sensor region of EV-A71 and CVA10, respectively, which can also facilitate viral entry [137, 140]. hSCARB2 also induces EV-A71 uncoating under acidic conditions [140,141,142]. Additionally, the human tryptophan-tRNA synthetase (hWARS) induced by interferon (IFN)-γ has also been recognized as a crucial factor in the entry of EVs [143]. The diversity of receptors and various modes of binding promote EVs infection.

Table 2 Major receptors for EVs

Human intestinal cells permit infection by EVs such as CVB3 and EV-A71, and can facilitate their replication and release [144]. EV-A71 infects the intestinal epithelium through the apical surface, with a preference for infecting goblet cells. hSCARB2, expressed as an integral membrane protein in goblet cells and localized in intracellular vesicles, provides the necessary condition for viral infection [145]. Although intestinal epithelium induces type IFNs secretion to limit viral replication, viral infection reduces the expression of goblet cells-derived mucins, and alters goblet cell function [146]. Therefore, the targeting of goblet cells by EV-A71 for intestinal infection is likely driven by the enrichment of hSCARB2 in secretory vesicles within these cells, which exposes the receptor through apical mucus release [146]. It is possible that EVs attach to the apical surface using SA glycoproteins and SA-containing glycolipids with SA-linked glycans or dependent decay accelerating factors [147, 148]. Moreover, the tonsillar crypt squamous epithelium, which supports active viral replication, is also an important site for EV-A71 invasion and replication, and is an important source of viral shedding in blood [149]. EVs that invade host cells rapidly complete the viral life cycle (Fig. 2). Subsequently, the virus is released from host cells through a traditional cytolytic manner, and packaged within exosomes, which promote virus spread without causing cell lysis [150, 151]. EVs replicate profusely in cells at the initial site of infection, and then spread to adjacent lymphoid tissues, and next spread to the circulation and target tissues, eventually developing varying degrees of viremia [152]. The proportion of cases with HFMD suffering from viremia is correlated with the duration of complications. In patients with mild HFMD, viremia that occurs improves as symptoms diminish [153]. If viral replication and transmission are controlled at this stage, most infected children will be asymptomatic. However, higher viral loads lead to the development of HFMD as long as the viral infection in the host continues to develop [154]. Together, the virus replicates in the gut early in the infection, and then spreads to the spinal cord, brain, and muscles later in the infection [155]. A part of patients with HFMD develop into more serious complications, including encephalitis, aseptic meningitis, acute flaccid paralysis, and cardiopulmonary failure [156, 157]. The central nervous system (CNS) damage is very common in severe HFMD cases complicated with encephalitis, aseptic meningitis [156]. Clinical reports and animal necropsy studies related to HFMD have revealed the presence of EV antigens in neurons at various locations within the CNS. This suggests that the virus may invade the CNS by compromising the blood–brain barrier (BBB), traveling backwards along nerves, or hijacking immune cells as a means of transportation [152, 158, 159]. Among them, retrograde axonal transport is currently considered as the main pathway for the EVs to invade CNS [160, 161]. Ohka et al. have confirmed through experiments in microfluidic devices that hSCARB2 is necessary for the retrograde axonal transport of EV-A71 [162]. Autopsy pathology revealed significant perivascular intussusception, infiltration of inflammatory cells into the parenchymal, and microglial nodules in the affected CNS. This may have been caused by EVs entering the CNS and infecting neurons, glial cells, the brain stem, the dentate nucleus, and the hypothalamus, ultimately leading to nerve damage [159].

Innate immune evasion by EVs

The initial defense against virus is to activate the secretion of IFNs and other antiviral molecules at the site of infection, and to exert their antiviral effects through both autocrine and paracrine mechanisms. The host cell recognizes pathogen-associated molecular patterns (PAMPs) through three pathogen recognition receptors (PRRs): toll-like receptors (TLRs), retinoic acid-inducible gene (RIG-I)-like receptors (RLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) (Fig. 5) [163]. It was discovered that the TLR7, TLR3 and TLR9 can recognize the single-stranded RNA (ssRNA) and double-stranded RNA (dsRNA) of EVs, which then triggers the recruitment of the toll interlukin-1 receptor (TIR). These leads to the activation of the Toll/IL-1R domain-containing adapter-inducing IFN-β (TRIF), which in turn brings in MyD88 into endosomes to further activate innate immune response [164,165,166,167]. Other findings suggest that ssRNA and dsRNA are also recognized by the RLR, specifically through the interaction of RIG-I and melanoma differentiation-associated gene 5 (MDA5) with mitochondrial antiviral-signaling protein (MAVS) to activate TANK-binding kinase 1 (TBK1) /IKK-ɛ and IKK-α/β/γ. The phosphorylation of TBK1 activates interferon regulatory factor 3 (IRF3) and stimulates the transcription of IFNs genes [168]. NLRP3 (NOD-, LRR- and pyrin domain-containing 3), as a common inflammasome, has also been demonstrated to play a role in the innate immune response to EVs infection [169]. Additionally, some antiviral molecules could enhance the secretion of IFNs, such as ATP1B3, zinc-finger antiviral protein (ZAP) [170, 171]. There are other unknown pathways for EVs to activate the innate immune response. For example, RNA-binding proteins (RBPs) FUS/TLS inhibited viral replication by interacting with EVs RNA, mediating the formation of SGs and promoting the production of antiviral proinflammatory cytokines and IFN-I [172]. IFNs also directly exert antiviral effects and indirectly induce the transmembrane protein TMEM106A to interfere with the binding of viruses to receptors to reduce cell damage [173, 174].

Fig. 5
figure 5

Innate immune evasion by Enterovirus. ssRNA, dsRNA and various proteins of EVs during replication and translation can activate and escape innate immunity through different pathways. (1) Viral RNA is recognized by TLR3, TLR7, TLR8 and TLR9, and then activates TRAF, TRIF, MyD88 and their downstream linker molecules, causing phosphorylation of IRF3, IRF7 and NF-κB to translocate to the nucleus, and finally promote the secretion of interferons (IFNs). (2) 2A protease(pro) and 3Cpro are mainly recognized by RIG-I and MDA5, and bind to MAVS in mitochondria to activate TRAF3 and TRAF6. However, prior to signaling to IRF1, IRF3, and IRF7, host ncRNAs regulated by the virus target and inhibit the activation of TRAF, ultimately reducing IFNs production. (3) Binding of IFNs to the receptor IFNAR activates downstream JAK1 and Tyk2, which promotes the phosphorylation and translocation of STAT1 and STAT2 to the nucleus, initiating transcription of IFN-stimulated response elements (ISREs). However, this pathway is directly or indirectly inhibited by 3Cpro, 2Apro, and 2Bpro, resulting in decreased secretion of IFNs. (4) Assembly of the NLRP3 inflammasome requires the sensor NLRP3, the adaptor protein ASC, and pro-caspase-1. However, host-invading viruses can activate and inhibit the formation of the NLRP3 inflammatory complex. Solid line with arrows at the end indicates activation; dashed line with a small line at the end indicates inhibition; scissor indicates cutting

EVs have developed various tactics to suppress the antiviral response that is mediated by IFNs [163], and the primary impediment to the antiviral pathway is located prior to the production of IFNs. 2Apro and 3Cpro directly inhibit the production of IFNs and the expression of IFNs receptors [175]. EVs mainly act on a variety of protein molecules in the PRR signaling pathway to complete immune evasion. EVs mainly inhibit TLR-dependent signaling mainly by controlling the level of host non-coding RNA (ncRNA) to indirectly influence the TLRs’ ability to sense the host cell, as well as cleaving the downstream molecules MyD88 and TRIF to prevent the IFNs production[176, 177]. 3Cpro could also directly target those key proteins of TLR signaling pathway to inhibit IFNs production [178]. EVs primarily counteract the RLR signaling pathway mainly by directly or indirectly cleaving RIG-I and MDA5, and targeting downstream linker molecules, such as MAVs [179,180,181]. Current evidence supports the conclusion that EV-A71’s 3D RNA polymerase directly interacts with NLRP3 to form a “3D-NLRP3-ASC” ring structure, which promotes the assembly of the NLRP3 inflammasome complex and the secretion of IL-1β [169]. Meanwhile, EV-A71 2Apro and 3Cpro also cleave NLRP3 to counteract inflammasome activation and inhibit IL-1β secretion [182]. In addition, the IFNs and JAK/STAT signaling pathway are also a key step to further expand antiviral immunity [183]. However, EV-A71 2Bpro and 2Apro selectively target the interferon receptor (IFNAR) directly or indirectly, suppressing the nuclear translocation of STAT1/ STAT2 and the level of ISGE, ultimately limiting the performance of IFNs [184,185,186,187,188]. In addition to participating in common innate immune signaling pathways, EVs also directly inhibit antiviral protein molecules like ZAP and acyl-CoA oxidase 1 (ACOX1). They also indirectly target ubiquitinated key proteins, such as Ubc6e, to induce apoptosis and autophagy, which ultimately exacerbates viral infection [171, 189,190,191].

Adaptive immunity to EVs and HFMD

Adaptive immunity also evolved to provide a broader and more sophisticated recognition mechanism to eliminate viruses [192]. Clinical evidence suggests that EVs could elicit neutralizing antibody (NAb) against homotypic viruses [193]. The NAb titers in the serum samples of infected children, collected one day after the symptoms appeared, were more than three times higher than those in healthy children, with the peak occurring at second day [194]. The results of a study investigating the kinetics of EV-A71 NAb response in patients with HFMD showed that the NAb titers rapidly reached a peak within 2 weeks of onset and remained at high levels for 2 years [195]. The study have shown that the serum immunoglobulin IgM (g/L) level of neonatal patients complicated with encephalitis is significantly higher than that of neonatal with lower neurological score [196]. A significant increase in serum and spinal cord IgM and IgG was also observed in EV-A71-infected mice [167]. However, another study found that there is no significant difference between the NAb titers in serum of patients with different severity of HFMD [197]. However, further research is needed to determine the relationship between the antibody response and the severity of HFMD in patients. Chang et al. believe that it is the cellular immunity response rather than the humoral immunity that has a greater impact on determining the outcome of the EV-A71 infection [197]. The cellular immunity carried out by T cells is essential for maintaining body defense. The autopsy biopsy showed abnormal changes in CD4+T cells and CD8+T cells [198]. Lymphocyte subsets displayed in peripheral blood samples from children infected with EV-A71 showed a decrease in the total number of Th (helper T cell), Tc (killer T cell) and Treg cells (regulatory T cell), and an increase in the percentage of B cells, Th2 and Th17 cells [199]. Furthermore, Th1/Tc1 and Th17/Treg were significantly increased in children infected with mild and severe HFMD [200, 201]. The fast and efficient immune response is a solid line of defense against viral infection. However, an excessive and dysregulated immune response can trigger a series of chain reactions, mainly manifested as systemic immune dysregulation with a neurogenic component.

Cytokine profiles in HFMD

The levels of cytokines were found to be significantly different among healthy individuals, those with mild HFMD and those with severe HFMD, suggesting a critical role in the progression of the disease and providing potential targets for diagnosis and treatment [202]. Several studies have summarized the cytokines and chemokines associated with severe HFMD, including TNF-α, IFN-γ, IL-1β, IL-18, IL-33, IL -37, IL-4, IL-13, IL-6, IL-12, IL-23, IL-27, IL-35, IL-10, IL-22, IL-17F, IL-8, IP-10, MCP-1, G-CSF, and HMGB1 [202,203,204]. The innate cells that are involved in cytokine production include neutrophils, macrophages, and natural killer (NK) cells. Meanwhile, and adaptive cells that are involved in ‘cytokine storm’ are mainly various of subsets T cells [205, 206]. The systemic inflammation is usually associated with the breakdown of BBB that accompanies CNS injury, which leads to the entry of brain-derived proinflammatory cytokines into the circulation, further activating the inflammatory cascade including complement [207]. For example, among the lymphocyte chemokines detected, high levels of interferon-gamma-inducible protein-10 (IP-10) were found in the plasma and cerebral spinal fluid of patients with severe HFMD [208]. Additional experiments revealed that a deficiency in IP-10 significantly reduced the levels of Mig (monokine induced by IFN-γ) in serum, and levels of IFN-γ and the number of CD8+ T cells in the mouse brain, This, in turn, resulted in an increase mortality rate of EV-A71-infected mice [209]. In addition, the chemokine (C-X-C motif) ligand (CXCL)10 was dramatically upregulated in EV-positive meningoencephalitis group [210]. Our previous study suggested that CXCL10 was highly expressed in the vital organs (brain, lung, heart, and skeletal muscle) of CV-A2-infected mice. Further interference with the CXCL10/CXCR3 axis was found to reduce the levels of leukocytes, neutrophils, and macrophages in the organs of mice that were critically ill [211]. Critical HFMD patients showed a decreased in peripheral blood lymphocytes, a depletion of CD4+and CD8+T lymphocytes, and a decline cellular immunity [212]. Ultimately, the immune system collapses and multiple organs of the host are damaged, leading to irreversible multi-organ failure and death.

Mechanisms of neurological damage and cardiopulmonary failure

Fatal complications of infections affecting the nervous system are directly or indirectly linked to damage of nerve cells. EV-A71 3Cpro directly cleaves the host DNA repair enzyme poly (ADP-ribose) polymerase and induces apoptosis [213]. EV-A71 3Dpro indirectly induces apoptosis and inflammation by downregulates ACOX1 expression and promotes reactive oxygen species (ROS) generation [191, 214]. The CNS damage is not only related to viral replication, but frequently associated with immune activation [215]. Recent discoveries indicate that nerve cells that express TLR7, TLR3, TLR8, and TLR9 can rapidly induce the secretion of IFNs in response to infection with EVs, which provides antiviral protection [164,165,166,167]. For instance, EV-A71 triggers a response in glia cells that involves the production of Interleukin-12p40 through TLR9 signaling, leading to the generation of neurotoxic Inducible Nitric Oxide Synthase (iNOS)/Nitric Oxide (NO), resulting in encephalitis [216]. In addition, the Janus kinase (JAK)-signal transducer of activators of transcription (STAT) pathway also regulates the expression of IFNs [184, 187]. Conversely, the virus antagonizes the antiviral response of nerve cells by cleaving RIG-I, which further inhibits the JAK/STAT signaling pathway [217, 218].

PE is one of the most serious complications of HFMD aside for encephalitis, and is the primary reason of rapid death of patients with severe HFMD [212, 219]. The development of PE is closely linked to inflammation in the CNS and ‘cytokine storm’ that is triggered by abnormally high depletion of IL-10, IL-13, IFN-γ and a depletion of lymphocyte in plasma [207, 212]. The currently recognized pathogenesis of fulminant PE is neurogenic [219,220,221,222]. Autopsy results showed extensive inflammatory in the CNS with severe PE [223]. A clinical study in Taiwan showed a significant correlation between CNS involvement and PE in children infected with EV-A71 [219]. Similarly, acute PE caused by Japanese encephalitis is associated with disruption of the anti-hypertensive mechanisms in the medulla of the CNS [224]. In the development of HFMD, damage to CNS leads to immune disorders, which are primarily manifested by the excessive release of catecholamines and cytokines [8, 207, 212]. Wu et al. proposed that the further increase in pulmonary vascular permeability caused by inflammatory response is the underlying cause of PE [225]. The CVA6-infected mice and EV-A71-infected hSCARB2 KI mice exhibited significant PE and hemorrhage, with the infiltration of neutrophil and monocyte in brain and spinal cord [226, 227]. In CVA2-infected mouse model, endothelial dysfunction, local inflammation, and enhanced vascular permeability were confirmed to be involved in accelerating acute lung injury [228]. Cardiac damage caused by EVs mainly progresses to acute heart failure (AHF) and myocarditis. During the HFMD outbreak in Taiwan in 1997, some severe patients presented with AHF [229]. The main cause of AHF in patients is acute left ventricular dysfunction and regional wall motion abnormalities [230]. The underlying cause of cardiac damage may be hypercatecholamineremia caused by brainstem encephalitis, which further leads to the cardiotoxicity of AHF [231, 232]. Myocardial cell necrosis is rarely observed in cardiac autopsy of EV-A71-infected patients [221]. In recent years, the emerging CVB3 among children infected with EVs has garnered increased attention [233]. As the most common pathogen causing viral myocarditis, CVB3 seems to promote cardiac function damage mainly by inducing myocardial apoptosis and necrosis [234]. Despite Lucie et al. not providing a comprehensive explanation of a fatal case of CVA2-related myocarditis in France, our colleagues noticed significant inflammatory and swelling in the heart of a mouse model infected with CVA2 [235, 236].

HFMD treatment

Unfortunately, there are currently no established antiviral treatments for HFMD and no specific clinical management and treatment methods have been established. For common cases, general treatment is usually used, isolating patients to avoid cross-infection, and taking good oral and skin care to avoid contamination. According to the development of HFMD, the treatment corresponding to the intervention of critical patients usually includes antiviral therapy, immunoglobulin therapy, respiratory and circulatory system support, etc.

Antiviral therapy

IFN-α, and ribavirin treatment have shown positive effect in antiviral management of HFMD to some extent [237, 238]. Various drugs, like antiviral peptides, small molecules, have been identified promising candidates, but their full pre-clinical validation have yet to be reported [239, 240].

Intravenous immunoglobulin (IVIG)

In previous outbreak of HFMD, IVIG was used on a presumptive basis for the treatment of severe cases [223, 241,242,243]. Recently, some anecdotal evidence suggests that the use of IVIG in the early stage of HFMD can significantly improve the progression of the disease and reduce mortality [244, 245]. Compared with conventional therapy alone, conventional therapy combined with IVIG had shorter fever clearance time, shorter rash regression time, and shorter clinical cure time [246].

Respiratory support

Mechanical ventilation is the most effective treatment to improve oxygen supply to the body [247]. The application of indications and the withdrawal indications are described in Chinese guidelines for the diagnosis and treatment of HFMD (2018) [21]. If they occur seizures (frequent myoclonic jerks), routine anti-convulsant may be considered, such as sedation (e.g., midazolam) and/or anticonvulsants (e.g., phenytoin).

Treatment of catecholamine storm

Early application of esmolol can effectively stabilize the vital signs of severe HFMD by reducing serum catecholamine concentration, alleviating myocardial damage, improving cardiac function, and reducing inflammatory response. The phentolamine can reduce mortality and relieves the symptoms of EV-A71-induced PE, which is a potential therapeutic agent for neurogenic PE [248].

Cardiovascular support

Multiple inotropes to support cardiac function (milrinone, dobutamine, dopamine, epinephrine) have been applied in the clinical treatment [247]. Among them, Milrinone exhibits immunoregulatory and anti-inflammatory effects in the management of systemic inflammatory response in severe cases [249]. If the above drugs prove ineffective, vasopressin or levosimendan can be considered [21].

Intracranial pressure control

Mannitol is commonly used to reduce increased intracranial pressure, the combination with hypertonic saline or diuretics may be considered for patients with severe intracranial hypertension [21, 250].

Traditional Chinese medicine

The combined Chinese medicine and chemistry medicine therapy achieve a better therapeutic efficacy in the treatment of severe HFMD than the chemistry medicine therapy alone [251]. The addition of Andrographolide Sulfonate and S. baicalensis to conventional therapy also reduces the occurrence of major complications, relieves fever, and attenuates oral lesions and rashes [252, 253].

Others

A retrospective observational study showed that continuous veno-venous hemodiafiltration could improve cardiovascular function [254]. Extracorporeal life support, including extracorporeal membrane oxygenation (ECMO), is last rescue treatment for patients who have failed to routine symptomatic and supportive treatment [8, 21].

Taken together, the main approach to treating severe cases of HFMD is mainly through supportive and symptom-relieving measures. There is a need to carry out more clinical studies to gather more evidence to improve the clinical management of severe cases.

Long-term sequelae of HFMD

Severe HFMD occurs mainly affects preschool children under the age of 5, a crucial stage in their growth and development. Although treatment advancements have led to a decrease in acute mortality, there are still concerns about the potential possible short-term or long-term impacts (Fig. 1).

Neurological dysfunction

A substantial burden of neurological sequelae following HFMD has been given more attention, especially in severe cases [6, 255, 256]. Among patients who experienced cardiopulmonary failure after CNS involvement, the proportion with subsequent sequelae (facial nerve palsy, limb weakness and atrophy, dysphagia, central hypoventilation, seizure, and psychomotor retardation) was significantly higher compared to those who only CNS involvement. The clinical severity of CNS involvement was significantly related to the children’s neurodevelopment (a delay in the gross motor and personal-social categories, delayed neurodevelopment) [257,258,259,260]. Serious virus-associated CNS infection during childhood appear to be associated with the later mental disorders, like attention-deficit hyperactivity disorder (ADHD) diagnosis alongside social/communication/emotion problems and autistic features [261,262,263]. Some severe EV-A71 infected patients may experience impaired speech and language skills due to subcortical white matter involvement in the acute stage [258,259,260]. Long-term functional neurological morbidity is associated with the involvement of medulla oblongata, gray matter in the brainstem or spinal cord, which may be closely monitored for early intervention and meticulous management [258, 264, 265].

Visual impairment

HFMD-related eye involvement presents variable signs, including pseudomembranous conjunctivitis [266], outer retinitis [267] and maculopathy [268], which is only observed in young adult patients in both sexes and always unilaterally. Despite self-limited nature and complete visual recovery in most cases later than resolution of HFMD symptoms (several weeks to months), some cases may have residual visual loss.

Nail abnormalities

Delayed skin and nail change, such as desquamation of palms and soles [269, 270], Beau’s lines, or onychomadesis [271], have also been observed in some severe EV-A71 infected patients. Nail change, mainly presenting as onychomadesis involving toenails or fingernails, is usually observed among 1–2 months after the onset of HFMD and lasted for 1–8 weeks, most for approximately 4 weeks and the changes are more likely to occur synchronously [272]. It can occur in both children and adults [273, 274]. The pathogens associated with nail abnormalities in HFMD patients are various, but mainly caused by CVA6 [269]. Nail change is usually self-limited with spontaneous healed requiring no treatment for all patients [275, 276].

In addition to focusing on the common health effects of HFMD, other health problems should not be ignored. Allergic diseases: a population-based cohort study has revealed that children suffered from HFMD had decreased risks of asthma [277]. In contrast, another retrospective cohort study found that the risk of asthma was higher in children with herpangina and HFMD [278]. Diabetes: One adult patient with severe atypical HFMD associated with CVA6 viremia showed impaired glucose tolerance after 2-year follow-up [279]. Heart diseases: A population-based cohort study has showed meningitis caused by herpangina/HFMD is the main disease associated with a higher risk of Kawasaki disease [280]. Idiopathic ventricular tachycardia, degenerative aortic valve disease, degenerative mitral valve disease, may be considered as sequelae of CVA6 infection in adults [279]. Nephropathy: A large national cohort study showed that children infected with EVs, particularly coxsackieviruses, had a significantly increased risk of developing nephrotic syndrome [281]. Leukemia: The risk of leukemia was significantly lower in the EVs-infected cohort, and herpangina/HFMD was the main disease reduced the risk of leukemia [282]. Long-term follow-up programs are crucial for early recognition of possible sequelae and early intervention in children who have suffered from HFMD, especially at a young age. Further studies are needed to better understand the pathogenesis of HFMD and its impact on sequelae.

Vaccine development

Vaccination is considered the most effective and cost-effective approach to control the incidence of HFMD. Currently, there are monovalent and polyvalent vaccines available against the HFMD pathogen. The monovalent vaccines consist mainly of inactivated whole virus vaccines, synthetic peptide and protein vaccines [283], recombinant subunit vaccines [284], and recombinant virus-vector vaccine [285]. Currently, the most readily available inactivated whole virus vaccines for EV71 are produced by Sinovac, Vigo, and the Chinese Academy of Medical Sciences (CAMS). Results from a randomised, double-blind phase 3 trial in China showed that the inactivated EV71 vaccine has a 97.4% efficacy rate [286]. The monovalent inactivated virus vaccine candidates for CVA16, CVA10, CVA6, and CVA5 have only been studied in animal models and lack clinical evidence of protection [287,288,289,290]. However, the limited scope of protection offered by monovalent vaccines, which are specific to one genotype, means that they do not provide protection against other EVs-associated cases of HFMD. Therefore, the most effective approach for reducing the incidence of HFMD is to use polyvalent vaccines that have been developed through the combination of effective monovalent vaccines or by constructing chimeric vaccines with different virus serotypes, which can provide better cross-reactivity and protection. Polyvalent vaccines, which aim to improve cross-reactivity, consist mainly of inactivated polyvalent vaccines, polyvalent virus-like particle vaccines, innovative chimeric vaccines, and recombinant virus-vector vaccines. Currently, the inactivated polyvalent vaccines, including bivalent, trivalent, and quadrivalent vaccines, have mainly been tested for their protective effects in animal studies. Vaccines formulated by combining inactivated EV-A71 and CVA16 viruses induced specific immunity against EV-A71 and CVA16 infections in animal models [291, 292]. The CVA6 and CVA10 inactivated whole virus bivalent vaccines have been shown to elicit high levels of neutralizing antibodies in mice [293]. The induction of a strong neutralizing antibody response and cell-mediated immune response was also shown to occur with the administration of inactivated whole virus trivalent vaccines [294, 295]. The antigen-specific and persistent serum antibody responses by quadrivalent vaccines were comparable to those by the respective monovalent vaccines [296]. In addition, polyvalent virus-like particles, novel chimeric vaccines, and recombinant virus-vector vaccines have all shown to induce broad protective effects and enhance systemic immune responses [297]. Antigenic peptide-based vaccine development and DNA/RNA vaccine technology be also applied for future exploration of polyvalent vaccines [298, 299]. However, it is important to carefully consider the inclusion of appropriate strains and to thoroughly evaluate the immunogenicity and immune interactions when developing multivalent vaccines.

Surveillance

The World Health Organization (WHO) primarily manages the existing global surveillance network for poliovirus, but has not yet established a specialized network to monitor HFMD or EVs. The National Enterovirus Surveillance System (NESS), established in the United States as a passive and laboratory-based system, has been used to track EVs reports since the 1960s, and provides the most comprehensive data for monitoring HFMD [300]. The Asia–Pacific Network for Enterovirus Surveillance (APNES) was established in 2017 through collaboration between academic institutions and hospitals in the Cambodia, Malaysia, Vietnam, and Taiwan region [301]. However, the efficiency of the system is limited due to its limited coverage and the absence of a unified governing body. In 2008, HFMD was incorporated into China’s notifiable infectious disease reporting system. In order to better prevent and control HFMD, China has gradually established and improved a nationwide monitoring network system for HFMD laboratories, with prefecture-level laboratories, provincial-level laboratories, and national-level laboratories as the main body. Most European countries have established national surveillance systems for laboratory-based detection of EVs. Currently, the Prospective, Multicenter and Cross-sectional Hospital Pilot Non-Polio Enterovirus Network program, which was jointly established by several European countries, is set to become operational in 2022 [302]. Laboratory-based disease surveillance networks can result in inefficient use of limited typing resources. Therefore, more optimized monitoring programs have been developed and applied to estimate HFMD incidence and optimize serotype estimation [303]. However, based on epidemiological data from dynamic surveillance of EVs that may cause HFMD, there has been a rise in the incidence of HFMD associated with some non-EV-A71/CVA EVs infections [304]. In recent years, the increasing occurrence of multiple EV infections and novel patterns of recombinant EV infections in patients with HFMD highlights the need for more vigilant pathogen surveillance of HFMD, especially in regards to emerging and co-infected pathogens [305].

Conclusions

In this Review, we systematically summarize the current knowledge on virology, epidemiology, pathogenesis, long-term sequelae of HFMD. Finally, as we assemble and interpret this evolving knowledge base, we need to understand which approaches to prevention and treatment, in this context, are most feasible and cost-effective, requiring a concerted effort between basic medical researchers and pediatricians. Overall, our study provides all relevant knowledge and the latest progress of HFMD, which will better inform health care and policy.

Availability of data and materials

All relevant data are within the manuscript and its additional files.

Abbreviations

HFMD:

Hand-foot-and-mouth disease

CFDA:

China Food and Drug Administration

CV:

Coxsackievirus

Echo:

Echoviruses

EVs:

Enteroviruses

PV:

Poliovirus

ORF:

Open reading frame

PE:

Pulmonary edema

COVID-19:

Coronavirus disease

hSCARB2:

Human scavenger receptor class B member 2

PSGL-1:

P-selectin glycoprotein ligand-1

BBB:

Blood–brain barrier

PAMPs:

Pathogen-associated molecular patterns

PRRs:

Pathogen recognition receptors

TLRs:

Toll-like receptors

RIG-I:

Retinoic acid-inducible gene

RLRs:

RIG-I-like receptors

NOD:

Nucleotide-binding oligomerization domain

NLRs:

NOD-like receptors

ssRNA:

Single-stranded RNA

dsRNA:

Double-stranded RNA

TIR:

The toll interlukin-1 receptor

TRIF:

The Toll/IL-1R domain-containing adapter-inducing IFN-β

MDA5:

Melanoma differentiation-associated gene 5

MAVS:

Mitochondrial antiviral-signaling protein

TBK1:

TANK-binding kinase 1

IRF3:

Interferon regulatory factor 3

ZAP:

Zinc-finger antiviral protein

RBPs:

RNA-binding proteins

ncRNA:

Non-coding RNA

IFNAR:

IFNs receptor

ACOX1:

Acyl-CoA oxidase 1

NAb:

Neutralizing antibody

NK:

Natural killer

IP-10:

Interferon-gamma-inducible protein-10

CXCL:

The chemokine ligand

ROS:

Reactive oxygen species

JAK:

Janus kinase

STAT:

Signal transducer of activators of transcription

AHF:

Acute heart failure

IVIG:

Intravenous immunoglobulin

ECMO:

Extracorporeal membrane oxygenation

ADHD:

Attention-deficit hyperactivity disorder

CAMS:

The Chinese Academy of Medical Sciences

NESS:

National Enterovirus Surveillance System

WHO:

The World Health Organization

APNES:

The Asia–Pacific Network for Enterovirus Surveillance

References

  1. Robinson CR, Doane FW, Rhodes AJ. Report of an outbreak of febrile illness with pharyngeal lesions and exanthem: Toronto, summer 1957; isolation of group A Coxsackie virus. Can Med Assoc J. 1958;79(8):615–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Clarke M, Hunter M, Mc NG, Von Seydlitz D, Rhodes AJ. Seasonal aseptic meningitis caused by Coxsackie and ECHO viruses, Toronto, 1957. Can Med Assoc J. 1959;81(1):5–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Alsop J, Flewett TH, Foster JR. “Hand-foot-and-mouth disease” in Birmingham in 1959. Br Med J. 1960;2(5214):1708–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Bubba L, Broberg EK, Jasir A, Simmonds P, Harvala H. Circulation of non-polio enteroviruses in 24 EU and EEA countries between 2015 and 2017: a retrospective surveillance study. Lancet Infect Dis. 2020;20(3):350–61.

    Article  PubMed  Google Scholar 

  5. Schmidt NJ, Lennette EH, Ho HH. An apparently new enterovirus isolated from patients with disease of the central nervous system. J Infect Dis. 1974;129(3):304–9.

    Article  CAS  PubMed  Google Scholar 

  6. Chang LY, Lin HY, Gau SS, Lu CY, Hsia SH, Huang YC, Huang LM, Lin TY. Enterovirus A71 neurologic complications and long-term sequelae. J Biomed Sci. 2019;26(1):57.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Gonzalez G, Carr MJ, Kobayashi M, Hanaoka N, Fujimoto T. Enterovirus-associated hand-foot and mouth disease and neurological complications in Japan and the rest of the world. Int J Mol Sci. 2019;20(20).

  8. Hsia SH, Lin JJ, Chan OW, Lin TY. Cardiopulmonary failure in children infected with Enterovirus A71. J Biomed Sci. 2020;27(1):53.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Chavda V.P., Patel K. and Apostolopoulos V. Tomato flu outbreak in India. Lancet Respir Med. 2022.

  10. Solomon T, Lewthwaite P, Perera D, Cardosa MJ, McMinn P, Ooi MH. Virology, epidemiology, pathogenesis, and control of enterovirus 71. Lancet Infect Dis. 2010;10(11):778–90.

    Article  PubMed  Google Scholar 

  11. Huang J, Liao Q, Ooi MH, Cowling BJ, Chang Z, Wu P, Liu F, Li Y, Luo L, Yu S, Yu H, Wei S. Epidemiology of recurrent hand, foot and mouth disease, China, 2008–2015. Emerg Infect Dis. 2018; 24(3).

  12. Hoang MTV, Nguyen TA, Tran TT, Vu TTH, Le NTN, Nguyen THN, Le THN, Nguyen TTH, Nguyen TH, Le NTN, Truong HK, Du TQ, Ha MT, Ho LV, Do CV, Nguyen TN, Nguyen TMT, Sabanathan S, Phan TQ, Van Nguyen VC, Thwaites GE, Wills B, Thwaites CL, Le VT, van Doorn HR. Clinical and aetiological study of hand, foot and mouth disease in southern Vietnam, 2013–2015: inpatients and outpatients. Int J Infect Dis IJID. 2019;80:1–9.

    Article  PubMed  Google Scholar 

  13. Zhao TS, Du J, Sun DP, Zhu QR, Chen LY, Ye C, Wang S, Liu YQ, Cui F, Lu QB. A review and meta-analysis of the epidemiology and clinical presentation of coxsackievirus A6 causing hand-foot-mouth disease in China and global implications. Rev Med Virol. 2020;30(2): e2087.

    Article  PubMed  Google Scholar 

  14. Gopalkrishna V, Patil PR, Patil GP, Chitambar SD. Circulation of multiple enterovirus serotypes causing hand, foot and mouth disease in India. J Med Microbiol. 2012;61(Pt 3):420–5.

    Article  CAS  PubMed  Google Scholar 

  15. Yao X, Bian LL, Lu WW, Li JX, Mao QY, Wang YP, Gao F, Wu X, Ye Q, Li XL, Zhu FC, Liang Z. Epidemiological and etiological characteristics of herpangina and hand foot mouth diseases in Jiangsu, China, 2013–2014. Hum Vaccin Immunother. 2017;13(4):823–30.

    Article  PubMed  Google Scholar 

  16. Plevka P, Perera R, Cardosa J, Kuhn RJ, Rossmann MG. Crystal structure of human enterovirus 71. Science. 2012;336(6086):1274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. McMinn PC. An overview of the evolution of enterovirus 71 and its clinical and public health significance. FEMS Microbiol Rev. 2002;26(1):91–107.

    Article  CAS  PubMed  Google Scholar 

  18. Chen J, Zhang C, Zhou Y, Zhang X, Shen C, Ye X, Jiang W, Huang Z, Cong Y. A 3.0-Angstrom resolution cryo-electron microscopy structure and antigenic sites of coxsackievirus A6-like particles. J Virol. 2018;92(2).

  19. Huang SC, Hsu YW, Wang HC, Huang SW, Kiang D, Tsai HP, Wang SM, Liu CC, Lin KH, Su IJ, Wang JR. Appearance of intratypic recombination of enterovirus 71 in Taiwan from 2002 to 2005. Virus Res. 2008;131(2):250–9.

    Article  CAS  PubMed  Google Scholar 

  20. Fu X, Wan Z, Li Y, Hu Y, Jin X, Zhang C. National epidemiology and evolutionary history of four hand, foot and mouth disease-related enteroviruses in China from 2008 to 2016. Virol Sin. 2020;35(1):21–33.

    Article  CAS  PubMed  Google Scholar 

  21. Li XW, Ni X, Qian SY, Wang Q, Jiang RM, Xu WB, Zhang YC, Yu GJ, Chen Q, Shang YX, Zhao CS, Yu H, Zhang T, Liu G, Deng HL, Gao J, Ran XG, Yang QZ, Xu BL, Huang XY, Wu XD, Bao YX, Chen YP, Chen ZH, Liu QQ, Lu GP, Liu CF, Wang RB, Zhang GL, Gu F, Xu HM, Li Y, Yang T. Chinese guidelines for the diagnosis and treatment of hand, foot and mouth disease (2018 edition). World J Pediatr. 2018;14(5):437–47.

    Article  PubMed  Google Scholar 

  22. Cox B, Levent F. Hand, foot, and mouth disease. JAMA. 2018;320(23):2492.

    Article  PubMed  Google Scholar 

  23. Fang Y, Wang S, Zhang L, Guo Z, Huang Z, Tu C, Zhu BP. Risk factors of severe hand, foot and mouth disease: a meta-analysis. Scand J Infect Dis. 2014;46(7):515–22.

    Article  PubMed  Google Scholar 

  24. Xing W, Liao Q, Viboud C, Zhang J, Sun J, Wu JT, Chang Z, Liu F, Fang VJ, Zheng Y, Cowling BJ, Varma JK, Farrar JJ, Leung GM, Yu H. Hand, foot, and mouth disease in China, 2008–12: an epidemiological study. Lancet Infect Dis. 2014;14(4):308–18.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Hoorn B, Tyrrell DA. On the growth of certain “newer” respiratory viruses in organ cultures. Br J Exp Pathol. 1965;46:109–18.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Wang Y, Zhao H, Ou R, Zhu H, Gan L, Zeng Z, Yuan R, Yu H, Ye M. Epidemiological and clinical characteristics of severe hand-foot-and-mouth disease (HFMD) among children: a 6-year population-based study. BMC Public Health. 2020;20(1):801.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Ben-Chetrit E, Wiener-Well Y, Shulman LM, Cohen MJ, Elinav H, Sofer D, Feldman I, Marva E, Wolf DG. Coxsackievirus A6-related hand foot and mouth disease: skin manifestations in a cluster of adult patients. J Clin Virol. 2014;59(3):201–3.

    Article  PubMed  Google Scholar 

  28. Li P, Rui J, Niu Y, Xie F, Wang Y, Li Z, Liu C, Yu S, Huang J, Luo L, Deng B, Liu W, Yang T, Li Q, Chen T. Analysis of HFMD transmissibility among the whole population and age groups in a large City of China. Front Public Health. 2022;10: 850369.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Zhao Z, Zheng C, Qi H, Chen Y, Ward MP, Liu F, Hong J, Su Q, Huang J, Chen X, Le J, Liu X, Ren M, Ba J, Zhang Z, Chang Z, Li Z. Impact of the coronavirus disease 2019 interventions on the incidence of hand, foot, and mouth disease in mainland China. Lancet Reg Health West Pac. 2022;20: 100362.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Shen L, Sun M, Song S, Hu Q, Wang N, Ou G, Guo Z, Du J, Shao Z, Bai Y, Liu K. The impact of anti-COVID-19 nonpharmaceutical interventions on hand, foot, and mouth disease-A spatiotemporal perspective in Xi’an, northwestern China. J Med Virol. 2022;94(7):3121–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Head JR, Collender PA, Lewnard JA, Skaff NK, Li L, Cheng Q, Baker JM, Li C, Chen D, Ohringer A, Liang S, Yang C, Hubbard A, Lopman B, Remais JV. Early evidence of inactivated enterovirus 71 vaccine impact against hand, foot, and mouth disease in a major center of ongoing transmission in China, 2011–2018: a longitudinal surveillance study. Clin Infect Dis. 2020;71(12):3088–95.

    Article  PubMed  Google Scholar 

  32. Esposito S, Principi N. Hand, foot and mouth disease: current knowledge on clinical manifestations, epidemiology, aetiology and prevention. Eur J Clin Microbiol Infect Dis. 2018;37(3):391–8.

    Article  PubMed  Google Scholar 

  33. Zhang R, Lin Z, Guo Z, Chang Z, Niu R, Wang Y, Wang S, Li Y. Daily mean temperature and HFMD: risk assessment and attributable fraction identification in Ningbo China. J Expo Sci Environ Epidemiol. 2021;31(4):664–71.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Bian L, Wang Y, Yao X, Mao Q, Xu M, Liang Z. Coxsackievirus A6: a new emerging pathogen causing hand, foot and mouth disease outbreaks worldwide. Expert Rev Anti Infect Ther. 2015;13(9):1061–71.

    Article  CAS  PubMed  Google Scholar 

  35. Rui J, Luo K, Chen Q, Zhang D, Zhao Q, Zhang Y, Zhai X, Zhao Z, Zhang S, Liao Y, Hu S, Gao L, Lei Z, Wang M, Wang Y, Liu X, Yu S, Xie F, Li J, Liu R, Chiang YC, Zhao B, Su Y, Zhang XS, Chen T. Early warning of hand, foot, and mouth disease transmission: a modeling study in mainland, China. PLoS Negl Trop Dis. 2021;15(3): e0009233.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Cheng Q, Bai L, Zhang Y, Zhang H, Wang S, Xie M, Zhao D, Su H. Ambient temperature, humidity and hand, foot, and mouth disease: a systematic review and meta-analysis. Sci Total Environ. 2018;625:828–36.

    Article  CAS  PubMed  Google Scholar 

  37. Bo Z, Ma Y, Chang Z, Zhang T, Liu F, Zhao X, Long L, Yi X, Xiao X, Li Z. The spatial heterogeneity of the associations between relative humidity and pediatric hand, foot and mouth disease: evidence from a nation-wide multicity study from mainland China. Sci Total Environ. 2020;707: 136103.

    Article  CAS  PubMed  Google Scholar 

  38. Gao Y, Wang H, Yi S, Wang D, Ma C, Tan B, Wei Y. Spatial and temporal characteristics of hand-foot-and-mouth disease and their influencing factors in Urumqi, China. Int J Environ Res Public Health. 2021;18(9):4919.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Zhang D, Li Z, Zhang W, Guo P, Ma Z, Chen Q, Du S, Peng J, Deng Y, Hao Y. Hand-washing: the main strategy for avoiding hand, foot and mouth disease. Int J Environ Res Public Health. 2016;13(6):610.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Guo N, Ma H, Deng J, Ma Y, Huang L, Guo R, Zhang L. Effect of hand washing and personal hygiene on hand food mouth disease: a community intervention study. Medicine (Baltimore). 2018;97(51): e13144.

    Article  PubMed  Google Scholar 

  41. Chen S, Yi K, Chen X, Li L, Tan X. A simple scoring system for quick, accurate, and reliable early diagnosis of hand, foot, and mouth disease. Med Sci Monit. 2018;24:8627–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lin H, Sun L, Lin J, He J, Deng A, Kang M, Zeng H, Ma W, Zhang Y. Protective effect of exclusive breastfeeding against hand, foot and mouth disease. BMC Infect Dis. 2014;14:645.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Upala P, Apidechkul T, Suttana W, Kullawong N, Tamornpark R, Inta C. Molecular epidemiology and clinical features of hand, foot and mouth disease in northern Thailand in 2016: a prospective cohort study. BMC Infect Dis. 2018;18(1):630.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Xie C, Wen H, Yang W, Cai J, Zhang P, Wu R, Li M, Huang S. Trend analysis and forecast of daily reported incidence of hand, foot and mouth disease in Hubei, China by Prophet model. Sci Rep. 2021;11(1):1445.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Deibel R, Gross LL, Collins DN. Isolation of a new enterovirus (38506). Proc Soc Exp Biol Med. 1975;148(1):203–7.

    Article  CAS  PubMed  Google Scholar 

  46. Melnick JL. Enterovirus type 71 infections: a varied clinical pattern sometimes mimicking paralytic poliomyelitis. Rev Infect Dis. 1984;6(Suppl 2):S387-390.

    Article  PubMed  Google Scholar 

  47. Bible JM, Iturriza-Gomara M, Megson B, Brown D, Pantelidis P, Earl P, Bendig J, Tong CY. Molecular epidemiology of human enterovirus 71 in the United Kingdom from 1998 to 2006. J Clin Microbiol. 2008;46(10):3192–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Shindarov LM, Chumakov MP, Voroshilova MK, Bojinov S, Vasilenko SM, Iordanov I, Kirov ID, Kamenov E, Leshchinskaya EV, Mitov G, Robinson IA, Sivchev S, Staikov S. Epidemiological, clinical, and pathomorphological characteristics of epidemic poliomyelitis-like disease caused by enterovirus 71. J Hyg Epidemiol Microbiol Immunol. 1979;23(3):284–95.

    CAS  PubMed  Google Scholar 

  49. Iacazio G, Martini D, Faure B, N’Guyen MH. Isolation and characterisation of 8-hydroxy-3Z,5Z-tetradecadienoic acid, a putative intermediate in Pichia guilliermondii gamma-decalactone biosynthesis from ricinoleic acid. FEMS Microbiol Lett. 2002;209(1):57–62.

    Article  CAS  PubMed  Google Scholar 

  50. Nagy G, Takatsy S, Kukan E, Mihaly I, Domok I. Virological diagnosis of enterovirus type 71 infections: experiences gained during an epidemic of acute CNS diseases in Hungary in 1978. Adv Virol. 1982;71(3):217–27.

    CAS  Google Scholar 

  51. van der Sanden S, Koopmans M, Uslu G, van der Avoort H, Dutch Working Group for Clinical V. Epidemiology of enterovirus 71 in the Netherlands, 1963 to 2008. J Clin Microbiol. 2009;47(9):2826–33.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ishimaru Y, Nakano S, Yamaoka K, Takami S. Outbreaks of hand, foot, and mouth disease by enterovirus 71. High incidence of complication disorders of central nervous system. Arch Dis Child. 1980;55(8):583–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Takimoto S, Waldman EA, Moreira RC, Kok F, Pinheiro Fde P, Saes SG, Hatch M, de Souza DF, Carmona Rde C, Shout D, de Moraes JC, Costa AM. Enterovirus 71 infection and acute neurological disease among children in Brazil (1988–1990). Trans R Soc Trop Med Hyg. 1998;92(1):25–8.

    Article  CAS  PubMed  Google Scholar 

  54. Chua KB, Kasri AR. Hand foot and mouth disease due to enterovirus 71 in Malaysia. Virol Sin. 2011;26(4):221–8.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Liu CC, Tseng HW, Wang SM, Wang JR, Su IJ. An outbreak of enterovirus 71 infection in Taiwan, 1998: epidemiologic and clinical manifestations. J Clin Virol. 2000;17(1):23–30.

    Article  CAS  PubMed  Google Scholar 

  56. Donato C, le Hoi T, Hoa NT, Hoa TM, Van Duyet L, Dieu Ngan TT, Van Kinh N, Vu Trung N, Vijaykrishna D. Genetic characterization of Enterovirus 71 strains circulating in Vietnam in 2012. Virology. 2016;495:1–9.

    Article  CAS  PubMed  Google Scholar 

  57. Akhmadishina LV, Govorukhina MV, Kovalev EV, Nenadskaya SA, Ivanova OE, Lukashev AN. Enterovirus A71 meningoencephalitis outbreak, Rostov-on-Don, Russia, 2013. Emerg Infect Dis. 2015;21(8):1440–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kim SJ, Kim JH, Kang JH, Kim DS, Kim KH, Kim KH, Kim YH, Chung JY, Bin JH, Jung DE, Kim JH, Kim HM, Cheon DS, Kang BH, Seo SY. Risk factors for neurologic complications of hand, foot and mouth disease in the Republic of Korea, 2009. J Korean Med Sci. 2013;28(1):120–7.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Shah VA, Chong CY, Chan KP, Ng W, Ling AE. Clinical characteristics of an outbreak of hand, foot and mouth disease in Singapore. Ann Acad Med Singap. 2003;32(3):381–7.

    CAS  PubMed  Google Scholar 

  60. Puenpa J, Auphimai C, Korkong S, Vongpunsawad S, Poovorawan Y. Enterovirus A71 infection, Thailand, 2017. Emerg Infect Dis. 2018;24(7):1386–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Noisumdaeng P, Korkusol A, Prasertsopon J, Sangsiriwut K, Chokephaibulkit K, Mungaomklang A, Thitithanyanont A, Buathong R, Guntapong R, Puthavathana P. Longitudinal study on enterovirus A71 and coxsackievirus A16 genotype/subgenotype replacements in hand, foot and mouth disease patients in Thailand, 2000–2017. Int J Infect Dis IJID. 2019;80:84–91.

    Article  PubMed  Google Scholar 

  62. Apostol LN, Suzuki A, Bautista A, Galang H, Paladin FJ, Fuji N, Lupisan S, Olveda R, Oshitani H. Detection of non-polio enteroviruses from 17 years of virological surveillance of acute flaccid paralysis in the Philippines. J Med Virol. 2012;84(4):624–31.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Fischer TK, Nielsen AY, Sydenham TV, Andersen PH, Andersen B, Midgley SE. Emergence of enterovirus 71 C4a in Denmark, 2009 to 2013. Euro Surveill. 2014;19(38).

  64. Luciani L, Morand A, Zandotti C, Piorkowski G, Boutin A, Mazenq J, Minodier P, Ninove L, Nougairede A. Circulation of enterovirus A71 during 2019–2020, Marseille, France. J Med Virol. 2021;93(8):5163–6.

    Article  CAS  PubMed  Google Scholar 

  65. Karrasch M, Fischer E, Scholten M, Sauerbrei A, Henke A, Renz DM, Mentzel HJ, Boer K, Bottcher S, Diedrich S, Krumbholz A, Zell R. A severe pediatric infection with a novel enterovirus A71 strain, Thuringia, Germany. J Clin Virol. 2016;84:90–5.

    Article  PubMed  Google Scholar 

  66. Gonzalez-Sanz R, Casas-Alba D, Launes C, Munoz-Almagro C, Ruiz-Garcia MM, Alonso M, Gonzalez-Abad MJ, Megias G, Rabella N, Del Cuerpo M, Gozalo-Marguello M, Gonzalez-Praetorius A, Martinez-Sapina A, Goyanes-Galan MJ, Romero MP, Calvo C, Anton A, Imaz M, Aranzamendi M, Hernandez-Rodriguez A, Moreno-Docon A, Rey-Cao S, Navascues A, Otero A, Cabrerizo M. Molecular epidemiology of an enterovirus A71 outbreak associated with severe neurological disease, Spain, 2016. Euro Surveill. 2019;24(7).

  67. Wieczorek M, Purzynska M, Krzysztoszek A, Ciacka A, Figas A, Szenborn L. Genetic characterization of enterovirus A71 isolates from severe neurological cases in Poland. J Med Virol. 2018;90(2):372–6.

    Article  PubMed  Google Scholar 

  68. Bendig JW, Fleming DM. Epidemiological, virological, and clinical features of an epidemic of hand, foot, and mouth disease in England and Wales. Commun Dis Rep CDR Rev. 1996;6(6):R81-86.

    CAS  PubMed  Google Scholar 

  69. Froeschle JE, Nahmias AJ, Feorino PM, McCord G, Naib Z. Hand, foot, and mouth disease (Coxsackievirus A16) in Atlanta. Am J Dis Child. 1967;114(3):278–83.

    CAS  PubMed  Google Scholar 

  70. Adler JL, Mostow SR, Mellin H, Janney JH, Joseph JM. Epidemiologic investigation of hand, foot, and mouth disease. Infection caused by coxsackievirus A 16 in Baltimore, June through September 1968. Am J Dis Child. 1970;120(4):309–14.

    Article  CAS  PubMed  Google Scholar 

  71. Ferson MJ, Bell SM. Outbreak of Coxsackievirus A16 hand, foot, and mouth disease in a child day-care center. Am J Public Health. 1991;81(12):1675–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zou XN, Zhang XZ, Wang B, Qiu YT. Etiologic and epidemiologic analysis of hand, foot, and mouth disease in Guangzhou city: a review of 4,753 cases. Braz J Infect Dis. 2012;16(5):457–65.

    Article  PubMed  Google Scholar 

  73. Zhu J, Luo Z, Wang J, Xu Z, Chen H, Fan D, Gao N, Ping G, Zhou Z, Zhang Y, An J. Phylogenetic analysis of Enterovirus 71 circulating in Beijing, China from 2007 to 2009. PLoS ONE. 2013;8(2): e56318.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Mori R, Kitahara Y, Takamori M. Studies of hand, foot and mouth disease. Virus isolation in an epidemic in Nobeoka city in 1970. Kansenshogaku Zasshi. 1971;45(3):105–11.

    Article  CAS  PubMed  Google Scholar 

  75. Kar BR, Dwibedi B, Kar SK. An outbreak of hand, foot and mouth disease in Bhubaneswar. Odisha Indian Pediatr. 2013;50(1):139–42.

    Article  PubMed  Google Scholar 

  76. Palani S, Nagarajan M, Biswas AK, Reesu R, Paluru V. Hand, foot and mouth disease in the Andaman Islands. India Indian Pediatr. 2018;55(5):408–10.

    Article  PubMed  Google Scholar 

  77. Chang LY. Enterovirus 71 in Taiwan. Pediatr Neonatol. 2008;49(4):103–12.

    Article  PubMed  Google Scholar 

  78. Van Tu P, Thao NTT, Perera D, Truong KH, Tien NTK, Thuong TC, How OM, Cardosa MJ, McMinn PC. Epidemiologic and virologic investigation of hand, foot, and mouth disease, southern Vietnam, 2005. Emerg Infect Dis. 2007;13(11):1733–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Goh KT, Doraisingham S, Tan JL, Lim GN, Chew SE. An outbreak of hand, foot, and mouth disease in Singapore. Bull World Health Organ. 1982;60(6):965–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Chan KP, Goh KT, Chong CY, Teo ES, Lau G, Ling AE. Epidemic hand, foot and mouth disease caused by human enterovirus 71, Singapore. Emerg Infect Dis. 2003;9(1):78–85.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Cabrerizo M, Tarrago D, Munoz-Almagro C, Del Amo E, Dominguez-Gil M, Eiros JM, Lopez-Miragaya I, Perez C, Reina J, Otero A, Gonzalez I, Echevarria JE, Trallero G. Molecular epidemiology of enterovirus 71, coxsackievirus A16 and A6 associated with hand, foot and mouth disease in Spain. Clin Microbiol Infect. 2014;20(3):O150-156.

    Article  CAS  PubMed  Google Scholar 

  82. Lizasoain A, Mir D, Martinez N, Colina R. Coxsackievirus A10 causing hand-foot-and-mouth disease in Uruguay. Int J Infect Dis IJID. 2020;94:1–3.

    Article  CAS  PubMed  Google Scholar 

  83. Nhan LNT, Khanh TH, Hong NTT, Van HMT, Nhu LNT, Ny NTH, Nguyet LA, Thanh TT, Anh NT, Hang VTT, Qui PT, Viet HL, Tung TH, Ha DQ, Tuan HM, Thwaites G, Chau NVV, Thwaites L, Hung NT, van Doorn HR, Tan LV. Clinical, etiological and epidemiological investigations of hand, foot and mouth disease in southern Vietnam during 2015–2018. PLoS Negl Trop Dis. 2020;14(8): e0008544.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Mirand A, Cohen R, Bisseux M, Tomba S, Sellem FC, Gelbert N, Bechet S, Frandji B, Archimbaud C, Brebion A, Chabrolles H, Regagnon C, Levy C, Bailly JL, Henquell C. A large-scale outbreak of hand, foot and mouth disease, France, as at 28 September 2021. Euro Surveill. 2021;26(43).

  85. Wang J, Zhou J, Xie G, Zheng S, Lou B, Chen Y, Wu Y. The epidemiological and clinical characteristics of hand, foot, and mouth disease in Hangzhou, China, 2016 to 2018. Clin Pediatr (Phila). 2020;59(7):656–62.

    Article  PubMed  Google Scholar 

  86. Blomqvist S, Klemola P, Kaijalainen S, Paananen A, Simonen ML, Vuorinen T, Roivainen M. Co-circulation of coxsackieviruses A6 and A10 in hand, foot and mouth disease outbreak in Finland. J Clin Virol. 2010;48(1):49–54.

    Article  CAS  PubMed  Google Scholar 

  87. Flett K, Youngster I, Huang J, McAdam A, Sandora TJ, Rennick M, Smole S, Rogers SL, Nix WA, Oberste MS, Gellis S, Ahmed AA. Hand, foot, and mouth disease caused by coxsackievirus a6. Emerg Infect Dis. 2012;18(10):1702–4.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Abedi GR, Watson JT, Pham H, Nix WA, Oberste MS, Gerber SI. Enterovirus and human parechovirus surveillance—United States, 2009–2013. MMWR Morb Mortal Wkly Rep. 2015;64(34):940–3.

    Article  PubMed  Google Scholar 

  89. Bujaki E, Farkas A, Rigo Z, Takacs M. Distribution of enterovirus genotypes detected in clinical samples in Hungary, 2010–2018. Acta Microbiol Immunol Hung. 2020;67(4):201–8.

    Article  PubMed  Google Scholar 

  90. Sinclair C, Gaunt E, Simmonds P, Broomfield D, Nwafor N, Wellington L, Templeton K, Willocks L, Schofield O, Harvala H. Atypical hand, foot, and mouth disease associated with coxsackievirus A6 infection, Edinburgh, United Kingdom, January to February 2014. Euro Surveill. 2014;19(12):20745.

    Article  CAS  PubMed  Google Scholar 

  91. Puenpa J, Chieochansin T, Linsuwanon P, Korkong S, Thongkomplew S, Vichaiwattana P, Theamboonlers A, Poovorawan Y. Hand, foot, and mouth disease caused by coxsackievirus A6, Thailand, 2012. Emerg Infect Dis. 2013;19(4):641–3.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Fujimoto T, Iizuka S, Enomoto M, Abe K, Yamashita K, Hanaoka N, Okabe N, Yoshida H, Yasui Y, Kobayashi M, Fujii Y, Tanaka H, Yamamoto M, Shimizu H. Hand, foot, and mouth disease caused by coxsackievirus A6, Japan, 2011. Emerg Infect Dis. 2012;18(2):337–9.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Kanbayashi D, Kaida A, Hirai Y, Yamamoto SP, Fujimori R, Okada M, Kubo H, Iritani N. An epidemic of hand, foot, and mouth disease caused by coxsackievirus A6 in Osaka City, Japan, in 2017. Jpn J Infect Dis. 2019;72(5):334–6.

    Article  CAS  PubMed  Google Scholar 

  94. Wu Y, Yeo A, Phoon MC, Tan EL, Poh CL, Quak SH, Chow VT. The largest outbreak of hand; foot and mouth disease in Singapore in 2008: the role of enterovirus 71 and coxsackievirus A strains. Int J Infect Dis. 2010;14(12):e1076-1081.

    Article  PubMed  Google Scholar 

  95. Hayman R, Shepherd M, Tarring C, Best E. Outbreak of variant hand-foot-and-mouth disease caused by coxsackievirus A6 in Auckland, New Zealand. J Paediatr Child Health. 2014;50(10):751–5.

    Article  PubMed  Google Scholar 

  96. Nelson BR, Edinur HA, Abdullah MT. Compendium of hand, foot and mouth disease data in Malaysia from years 2010–2017. Data Brief. 2019;24: 103868.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Li J, Zhu R, Huo D, Du Y, Yan Y, Liang Z, Luo Y, Yang Y, Jia L, Chen L, Wang Q, He Y. An outbreak of Coxsackievirus A6-associated hand, foot, and mouth disease in a kindergarten in Beijing in 2015. BMC Pediatr. 2018;18(1):277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Li Y, Chang Z, Wu P, Liao Q, Liu F, Zheng Y, Luo L, Zhou Y, Chen Q, Yu S, Guo C, Chen Z, Long L, Zhao S, Yang B, Yu H, Cowling BJ. Emerging enteroviruses causing hand, foot and mouth disease, China, 2010–2016. Emerg Infect Dis. 2018;24(10):1902–6.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Lau SKP, Zhao PSH, Sridhar S, Yip CCY, Aw-Yong KL, Chow EYY, Cheung KCM, Hui RWH, Leung RYH, Lai YSK, Wu AKL, To KKW, Woo PCY, Yuen KY. Molecular epidemiology of coxsackievirus A6 circulating in Hong Kong reveals common neurological manifestations and emergence of novel recombinant groups. J Clin Virol. 2018;108:43–9.

    Article  PubMed  Google Scholar 

  100. Hu L, Maimaiti H, Zhou L, Gao J, Lu Y. Changing serotypes of hand, foot and mouth disease in Shanghai, 2017–2019. Gut Pathog. 2022;14(1):12.

    Article  PubMed  PubMed Central  Google Scholar 

  101. Ramirez-Fort MK, Downing C, Doan HQ, Benoist F, Oberste MS, Khan F, Tyring SK. Coxsackievirus A6 associated hand, foot and mouth disease in adults: clinical presentation and review of the literature. J Clin Virol. 2014;60(4):381–6.

    Article  PubMed  Google Scholar 

  102. Hoang CQ, Nguyen HD, Ho NX, Vu THT, Pham TTM, Nguyen KT, Nguyen HT, Hoang LT, Clapham H, Nguyen TTT, Phan LT. Incidence of infection of enterovirus 71 and coxsackieviruses A6 and A16 among household contacts of index cases in Dong Thap Province, Southern Vietnam. Biomed Res Int. 2020;2020:9850351.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Oberste MS, Penaranda S, Maher K, Pallansch MA. Complete genome sequences of all members of the species Human enterovirus A. J Gen Virol. 2004;85(Pt 6):1597–607.

    Article  CAS  PubMed  Google Scholar 

  104. Duff MF. Hand-foot-and-mouth syndrome in humans: coxackie A10 infections in New Zealand. Br Med J. 1968;2(5606):661–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Itagaki A, Ishihara J, Mochida K, Ito Y, Saito K, Nishino Y, Koike S, Kurimura T. A clustering outbreak of hand, foot, and mouth disease caused by Coxsackie virus A10. Microbiol Immunol. 1983;27(11):929–35.

    Article  CAS  PubMed  Google Scholar 

  106. Mirand A, Henquell C, Archimbaud C, Ughetto S, Antona D, Bailly JL, Peigue-Lafeuille H. Outbreak of hand, foot and mouth disease/herpangina associated with coxsackievirus A6 and A10 infections in 2010, France: a large citywide, prospective observational study. Clin Microbiol Infect. 2012;18(5):E110-118.

    Article  CAS  PubMed  Google Scholar 

  107. Ryu WS, Kang B, Hong J, Hwang S, Kim J, Cheon DS. Clinical and etiological characteristics of enterovirus 71-related diseases during a recent 2-year period in Korea. J Clin Microbiol. 2010;48(7):2490–4.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Xie J, Yang XH, Hu SQ, Zhan WL, Zhang CB, Liu H, Zhao HY, Chai HY, Chen KY, Du QY, Liu P, Yin AH, Luo MY. Co-circulation of coxsackieviruses A-6, A-10, and A-16 causes hand, foot, and mouth disease in Guangzhou city, China. BMC Infect Dis. 2020;20(1):271.

    Article  PubMed  PubMed Central  Google Scholar 

  109. He SZ, Chen MY, Xu XR, Yan Q, Niu JJ, Wu WH, Su XS, Ge SX, Zhang SY, Xia NS. Epidemics and aetiology of hand, foot and mouth disease in Xiamen, China, from 2008 to 2015. Epidemiol Infect. 2017;145(9):1865–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Wang J, Liu J, Fang F, Wu J, Ji T, Yang Y, Liu L, Li C, Zhang W, Zhang X, Teng Z. Genomic surveillance of coxsackievirus A10 reveals genetic features and recent appearance of genogroup D in Shanghai, China, 2016–2020. Virol Sin. 2022;37(2):177–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lukashev AN, Vakulenko YA, Turbabina NA, Deviatkin AA, Drexler JF. Molecular epidemiology and phylogenetics of human enteroviruses: Is there a forest behind the trees? Rev Med Virol. 2018;28(6): e2002.

    Article  PubMed  Google Scholar 

  112. Brown BA, Oberste MS, Alexander JP Jr, Kennett ML, Pallansch MA. Molecular epidemiology and evolution of enterovirus 71 strains isolated from 1970 to 1998. J Virol. 1999;73(12):9969–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Zhang C, Zhu R, Yang Y, Chi Y, Yin J, Tang X, Yu L, Zhang C, Huang Z, Zhou D. Phylogenetic analysis of the major causative agents of hand, foot and mouth disease in Suzhou City, Jiangsu province, China, in 2012–2013. Emerg Microbes Infect. 2015;4(2): e12.

    PubMed  PubMed Central  Google Scholar 

  114. He Y, Zou L, Chong MKC, Men R, Xu W, Yang H, Yao X, Chen L, Xian H, Zhang H, Luo M, Cheng J, Ma H, Feng Q, Huang Y, Wang Y, Yeoh EK, Zee BC, Zhou Y, He ML, Wang MH. Genetic evolution of Human Enterovirus A71 subgenotype C4 in Shenzhen, China, 1998–2013. J Infect. 2016;72(6):731–7.

    Article  PubMed  Google Scholar 

  115. Saxena VK, Sane S, Nadkarni SS, Sharma DK, Deshpande JM. Genetic diversity of enterovirus A71, India. Emerg Infect Dis. 2015;21(1):123–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Fernandez-Garcia MD, Volle R, Joffret ML, Sadeuh-Mba SA, Gouandjika-Vasilache I, Kebe O, Wiley MR, Majumdar M, Simon-Loriere E, Sakuntabhai A, Palacios G, Martin J, Delpeyroux F, Ndiaye K, Bessaud M. Genetic characterization of enterovirus A71 circulating in Africa. Emerg Infect Dis. 2018;24(4):754–7.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Bessaud M, Razafindratsimandresy R, Nougairede A, Joffret ML, Deshpande JM, Dubot-Peres A, Heraud JM, de Lamballerie X, Delpeyroux F, Bailly JL. Molecular comparison and evolutionary analyses of VP1 nucleotide sequences of new African human enterovirus 71 isolates reveal a wide genetic diversity. PLoS ONE. 2014;9(3): e90624.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Nhu LNT, Nhan LNT, Anh NT, Hong NTT, Van HMT, Thanh TT, Hang VTT, Han DDK, Ny NTH, Nguyet LA, Quy DT, Qui PT, Khanh TH, Hung NT, Tuan HM, Chau NVV, Thwaites G, van Doorn HR, Tan LV. Coxsackievirus A16 in Southern Vietnam. Front Microbiol. 2021;12: 689658.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Hu YF, Jia LP, Yu FY, Liu LY, Song QW, Dong HJ, Deng J, Qian Y, Zhao LQ, Deng L, Huang H, Zhu RN. Molecular epidemiology of coxsackievirus A16 circulating in children in Beijing, China from 2010 to 2019. World J Pediatr. 2021;17(5):508–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Hassel C, Mirand A, Farkas A, Diedrich S, Huemer HP, Peigue-Lafeuille H, Archimbaud C, Henquell C, Bailly JL, Network H.F.S. Phylogeography of coxsackievirus A16 reveals global transmission pathways and recent emergence and spread of a recombinant genogroup. J Virol. 2017;91(18).

  121. Wang J, Teng Z, Chu W, Fang F, Cui X, Guo X, Zhang X, Thorley BR, Zhu Y. The emergence and spread of one Coxsackievirus A16 Genogroup D novel recombinant strain that caused a clustering HFMD outbreak in Shanghai, China, 2016. Emerg Microbes Infect. 2018;7(1):131.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Chen L, Yao XJ, Xu SJ, Yang H, Wu CL, Lu J, Xu WB, Zhang HL, Meng J, Zhang Y, He YQ, Zhang RL. Molecular surveillance of coxsackievirus A16 reveals the emergence of a new clade in mainland China. Adv Virol. 2019;164(3):867–74.

    CAS  Google Scholar 

  123. Song Y, Zhang Y, Ji T, Gu X, Yang Q, Zhu S, Xu W, Xu Y, Shi Y, Huang X, Li Q, Deng H, Wang X, Yan D, Yu W, Wang S, Yu D, Xu W. Persistent circulation of Coxsackievirus A6 of genotype D3 in mainland of China between 2008 and 2015. Sci Rep. 2017;7(1):5491.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Dalldorf G. The coxsackie virus group. Ann N Y Acad Sci. 1953;56(3):583–6.

    Article  CAS  PubMed  Google Scholar 

  125. Munivenkatappa A, Yadav PD, Nyayanit DA, Majumdar TD, Sangal L, Jain S, Sinha DP, Shrivastava A, Mourya DT. Molecular diversity of Coxsackievirus A10 circulating in the southern and northern region of India [2009-17]. Infect Genet Evol. 2018;66:101–10.

    Article  CAS  PubMed  Google Scholar 

  126. Lukashev AN, Shumilina EY, Belalov IS, Ivanova OE, Eremeeva TP, Reznik VI, Trotsenko OE, Drexler JF, Drosten C. Recombination strategies and evolutionary dynamics of the Human enterovirus A global gene pool. J Gen Virol. 2014;95(Pt 4):868–73.

    Article  CAS  PubMed  Google Scholar 

  127. Zhang M, Xu D, Feng C, Guo W, Fei C, Sun H, Yang Z, Ma S. Isolation and characterization of a novel clade of coxsackievirus B2 associated with hand, foot, and mouth disease in Southwest China. J Med Virol. 2022;94(6):2598–606.

    Article  CAS  PubMed  Google Scholar 

  128. Yang Q, Yan D, Song Y, Zhu S, He Y, Han Z, Wang D, Ji T, Zhang Y, Xu W. Whole-genome analysis of coxsackievirus B3 reflects its genetic diversity in China and worldwide. Virol J. 2022;19(1):69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Mendelsohn CL, Wimmer E, Racaniello VR. Cellular receptor for poliovirus: molecular cloning, nucleotide sequence, and expression of a new member of the immunoglobulin superfamily. Cell. 1989;56(5):855–65.

    Article  CAS  PubMed  Google Scholar 

  130. Ren RB, Costantini F, Gorgacz EJ, Lee JJ, Racaniello VR. Transgenic mice expressing a human poliovirus receptor: a new model for poliomyelitis. Cell. 1990;63(2):353–62.

    Article  CAS  PubMed  Google Scholar 

  131. Rossmann MG, He Y, Kuhn RJ. Picornavirus-receptor interactions. Trends Microbiol. 2002;10(7):324–31.

    Article  CAS  PubMed  Google Scholar 

  132. Yamayoshi S, Yamashita Y, Li J, Hanagata N, Minowa T, Takemura T, Koike S. Scavenger receptor B2 is a cellular receptor for enterovirus 71. Nat Med. 2009;15(7):798–801.

    Article  CAS  PubMed  Google Scholar 

  133. Nishimura Y, Shimojima M, Tano Y, Miyamura T, Wakita T, Shimizu H. Human P-selectin glycoprotein ligand-1 is a functional receptor for enterovirus 71. Nat Med. 2009;15(7):794–7.

    Article  CAS  PubMed  Google Scholar 

  134. Yang SL, Chou YT, Wu CN, Ho MS. Annexin II binds to capsid protein VP1 of enterovirus 71 and enhances viral infectivity. J Virol. 2011;85(22):11809–20.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Tan CW, Poh CL, Sam IC, Chan YF. Enterovirus 71 uses cell surface heparan sulfate glycosaminoglycan as an attachment receptor. J Virol. 2013;87(1):611–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Staring J, van den Hengel LG, Raaben M, Blomen VA, Carette JE, Brummelkamp TR. KREMEN1 is a host entry receptor for a major group of enteroviruses. Cell Host Microbe. 2018;23(5):636–43.

    Article  CAS  PubMed  Google Scholar 

  137. Zhao Y, Zhou D, Ni T, Karia D, Kotecha A, Wang X, Rao Z, Jones EY, Fry EE, Ren J, Stuart DI. Hand-foot-and-mouth disease virus receptor KREMEN1 binds the canyon of Coxsackie Virus A10. Nat Commun. 2020;11(1):38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Tuthill TJ, Groppelli E, Hogle JM, Rowlands DJ. Picornaviruses. Curr Top Microbiol Immunol. 2010;343:43–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Lin YW, Lin HY, Tsou YL, Chitra E, Hsiao KN, Shao HY, Liu CC, Sia C, Chong P, Chow YH. Human SCARB2-mediated entry and endocytosis of EV71. PLoS ONE. 2012;7(1): e30507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Zhou D, Zhao Y, Kotecha A, Fry EE, Kelly JT, Wang X, Rao Z, Rowlands DJ, Ren J, Stuart DI. Unexpected mode of engagement between enterovirus 71 and its receptor SCARB2. Nat Microbiol. 2019;4(3):414–9.

    Article  CAS  PubMed  Google Scholar 

  141. Wang X, Peng W, Ren J, Hu Z, Xu J, Lou Z, Li X, Yin W, Shen X, Porta C, Walter TS, Evans G, Axford D, Owen R, Rowlands DJ, Wang J, Stuart DI, Fry EE, Rao Z. A sensor-adaptor mechanism for enterovirus uncoating from structures of EV71. Nat Struct Mol Biol. 2012;19(4):424–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Dang M, Wang X, Wang Q, Wang Y, Lin J, Sun Y, Li X, Zhang L, Lou Z, Wang J, Rao Z. Molecular mechanism of SCARB2-mediated attachment and uncoating of EV71. Protein Cell. 2014;5(9):692–703.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Yeung ML, Jia L, Yip CCY, Chan JFW, Teng JLL, Chan KH, Cai JP, Zhang C, Zhang AJ, Wong WM, Kok KH, Lau SKP, Woo PCY, Lo JYC, Jin DY, Shih SR, Yuen KY. Human tryptophanyl-tRNA synthetase is an IFN-gamma-inducible entry factor for Enterovirus. J Clin Invest. 2018;128(11):5163–77.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Drummond CG, Bolock AM, Ma C, Luke CJ, Good M, Coyne CB. Enteroviruses infect human enteroids and induce antiviral signaling in a cell lineage-specific manner. Proc Natl Acad Sci USA. 2017;114(7):1672–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Yu P, Gao Z, Zong Y, Bao L, Xu L, Deng W, Li F, Lv Q, Gao Z, Xu Y, Yao Y, Qin C. Histopathological features and distribution of EV71 antigens and SCARB2 in human fatal cases and a mouse model of enterovirus 71 infection. Virus Res. 2014;189:121–32.

    Article  CAS  PubMed  Google Scholar 

  146. Good C, Wells AI, Coyne CB. Type III interferon signaling restricts enterovirus 71 infection of goblet cells. Sci Adv. 2019;5(3):eaau4255.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Shieh JT, Bergelson JM. Interaction with decay-accelerating factor facilitates coxsackievirus B infection of polarized epithelial cells. J Virol. 2002;76(18):9474–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Yang B, Chuang H, Yang KD. Sialylated glycans as receptor and inhibitor of enterovirus 71 infection to DLD-1 intestinal cells. Virol J. 2009;6:141.

    Article  PubMed  PubMed Central  Google Scholar 

  149. He Y, Ong KC, Gao Z, Zhao X, Anderson VM, McNutt MA, Wong KT, Lu M. Tonsillar crypt epithelium is an important extra-central nervous system site for viral replication in EV71 encephalomyelitis. Am J Pathol. 2014;184(3):714–20.

    Article  CAS  PubMed  Google Scholar 

  150. Gu J, Wu J, Fang D, Qiu Y, Zou X, Jia X, Yin Y, Shen L, Mao L. Exosomes cloak the virion to transmit Enterovirus 71 non-lytically. Virulence. 2020;11(1):32–8.

    Article  CAS  PubMed  Google Scholar 

  151. Wang Y, Zhang S, Song W, Zhang W, Li J, Li C, Qiu Y, Fang Y, Jiang Q, Li X, Yan B. Exosomes from EV71-infected oral epithelial cells can transfer miR-30a to promote EV71 infection. Oral Dis. 2020;26(4):778–88.

    Article  PubMed  Google Scholar 

  152. Zhang Y, Cui W, Liu L, Wang J, Zhao H, Liao Y, Na R, Dong C, Wang L, Xie Z, Gao J, Cui P, Zhang X, Li Q. Pathogenesis study of enterovirus 71 infection in rhesus monkeys. Lab Invest. 2011;91(9):1337–50.

    Article  PubMed  Google Scholar 

  153. Cheng HY, Huang YC, Yen TY, Hsia SH, Hsieh YC, Li CC, Chang LY, Huang LM. The correlation between the presence of viremia and clinical severity in patients with enterovirus 71 infection: a multi-center cohort study. BMC Infect Dis. 2014;14:417.

    Article  PubMed  PubMed Central  Google Scholar 

  154. Chang LY, King CC, Hsu KH, Ning HC, Tsao KC, Li CC, Huang YC, Shih SR, Chiou ST, Chen PY, Chang HJ, Lin TY. Risk factors of enterovirus 71 infection and associated hand, foot, and mouth disease/herpangina in children during an epidemic in Taiwan. Pediatrics. 2002;109(6): e88.

    Article  PubMed  Google Scholar 

  155. Wang YF, Chou CT, Lei HY, Liu CC, Wang SM, Yan JJ, Su IJ, Wang JR, Yeh TM, Chen SH, Yu CK. A mouse-adapted enterovirus 71 strain causes neurological disease in mice after oral infection. J Virol. 2004;78(15):7916–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Ho M, Chen ER, Hsu KH, Twu SJ, Chen KT, Tsai SF, Wang JR, Shih SR. An epidemic of enterovirus 71 infection in Taiwan. Taiwan Enterovirus Epidemic Working Group. N Engl J Med. 1999;341(13):929–35.

    Article  CAS  PubMed  Google Scholar 

  157. Lu MY, Lin YL, Kuo Y, Chuang CF, Wang JR, Liao F. Muscle tissue damage and recovery after EV71 infection correspond to dynamic macrophage phenotypes. Front Immunol. 2021;12: 648184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Hashimoto I, Hagiwara A. Pathogenicity of a poliomyelitis-like disease in monkeys infected orally with enterovirus 71: a model for human infection. Neuropathol Appl Neurobiol. 1982;8(2):149–56.

    Article  CAS  PubMed  Google Scholar 

  159. Wong KT, Munisamy B, Ong KC, Kojima H, Noriyo N, Chua KB, Ong BB, Nagashima K. The distribution of inflammation and virus in human enterovirus 71 encephalomyelitis suggests possible viral spread by neural pathways. J Neuropathol Exp Neurol. 2008;67(2):162–9.

    Article  PubMed  Google Scholar 

  160. Chen CS, Yao YC, Lin SC, Lee YP, Wang YF, Wang JR, Liu CC, Lei HY, Yu CK. Retrograde axonal transport: a major transmission route of enterovirus 71 in mice. J Virol. 2007;81(17):8996–9003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Pan H, Yao X, Chen W, Wang F, He H, Liu L, He Y, Chen J, Jiang P, Zhang R, Ma Y, Cai L. Dissecting complicated viral spreading of enterovirus 71 using in situ bioorthogonal fluorescent labeling. Biomaterials. 2018;181:199–209.

    Article  CAS  PubMed  Google Scholar 

  162. Ohka S, Hao Tan S, Kaneda S, Fujii T, Schiavo G. Retrograde axonal transport of poliovirus and EV71 in motor neurons. Biochem Biophys Res Commun. 2022;626:72–8.

    Article  CAS  PubMed  Google Scholar 

  163. Jin Y, Zhang R, Wu W, Duan G. Innate immunity evasion by enteroviruses linked to epidemic hand-foot-mouth disease. Front Microbiol. 2018;9:2422.

    Article  PubMed  PubMed Central  Google Scholar 

  164. Hsiao HB, Chou AH, Lin SI, Chen IH, Lien SP, Liu CC, Chong P, Liu SJ. Toll-like receptor 9-mediated protection of enterovirus 71 infection in mice is due to the release of danger-associated molecular patterns. J Virol. 2014;88(20):11658–70.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Chen KR, Yu CK, Kung SH, Chen SH, Chang CF, Ho TC, Lee YP, Chang HC, Huang LY, Lo SY, Chang JC, Ling P. Toll-like receptor 3 is involved in detection of enterovirus A71 infection and targeted by viral 2A protease. Viruses. 2018;10(12):689.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Luo Z, Su R, Wang W, Liang Y, Zeng X, Shereen MA, Bashir N, Zhang Q, Zhao L, Wu K, Liu Y, Wu J. EV71 infection induces neurodegeneration via activating TLR7 signaling and IL-6 production. PLoS Pathog. 2019;15(11): e1008142.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Lin YL, Lu MY, Chuang CF, Kuo Y, Lin HE, Li FA, Wang JR, Hsueh YP, Liao F. TLR7 is critical for anti-viral humoral immunity to EV71 infection in the spinal cord. Front Immunol. 2020;11: 614743.

    Article  CAS  PubMed  Google Scholar 

  168. Rasti M, Khanbabaei H, Teimoori A. An update on enterovirus 71 infection and interferon type I response. Rev Med Virol. 2019;29(1): e2016.

    Article  PubMed  Google Scholar 

  169. Gong Z, Gao X, Yang Q, Lun J, Xiao H, Zhong J, Cao H. Phosphorylation of ERK-dependent NF-kappaB triggers NLRP3 inflammasome mediated by vimentin in EV71-infected glioblastoma cells. Molecules. 2022;27(13):4190.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Lu Y, Hou H, Wang F, Qiao L, Wang X, Yu J, Liu W, Sun Z. ATP1B3: a virus-induced host factor against EV71 replication by up-regulating the production of type-I interferons. Virology. 2016;496:28–34.

    Article  CAS  PubMed  Google Scholar 

  171. Xie L, Lu B, Zheng Z, Miao Y, Liu Y, Zhang Y, Zheng C, Ke X, Hu Q, Wang H. The 3C protease of enterovirus A71 counteracts the activity of host zinc-finger antiviral protein (ZAP). J Gen Virol. 2018;99(1):73–85.

    Article  CAS  PubMed  Google Scholar 

  172. Xue YC, Ng CS, Mohamud Y, Fung G, Liu H, Bahreyni A, Zhang J, Luo H. FUS/TLS suppresses enterovirus replication and promotes antiviral innate immune responses. J Virol. 2021;95(12).

  173. Sen GC. Viruses and interferons. Annu Rev Microbiol. 2001;55:255–81.

    Article  CAS  PubMed  Google Scholar 

  174. Guo X, Zeng S, Ji X, Meng X, Lei N, Yang H, Mu X. Type I interferon-induced TMEM106A blocks attachment of EV-A71 virus by interacting with the membrane protein SCARB2. Front Immunol. 2022;13: 817835.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Dong Y, Liu J, Lu N, Zhang C. Enterovirus 71 antagonizes antiviral effects of type III interferon and evades the clearance of intestinal intraepithelial lymphocytes. Front Microbiol. 2021;12: 806084.

    Article  PubMed  Google Scholar 

  176. Ho BC, Yu IS, Lu LF, Rudensky A, Chen HY, Tsai CW, Chang YL, Wu CT, Chang LY, Shih SR, Lin SW, Lee CN, Yang PC, Yu SL. Inhibition of miR-146a prevents enterovirus-induced death by restoring the production of type I interferon. Nat Commun. 2014;5:3344.

    Article  PubMed  Google Scholar 

  177. Feng N, Zhou Z, Li Y, Zhao L, Xue Z, Lu R, Jia K. Enterovirus 71-induced has-miR-21 contributes to evasion of host immune system by targeting MyD88 and IRAK1. Virus Res. 2017;237:27–36.

    Article  CAS  PubMed  Google Scholar 

  178. Lei X, Sun Z, Liu X, Jin Q, He B, Wang J. Cleavage of the adaptor protein TRIF by enterovirus 71 3C inhibits antiviral responses mediated by Toll-like receptor 3. J Virol. 2011;85(17):8811–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Kuo RL, Kao LT, Lin SJ, Wang RY, Shih SR. MDA5 plays a crucial role in enterovirus 71 RNA-mediated IRF3 activation. PLoS ONE. 2013;8(5): e63431.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Rui Y, Su J, Wang H, Chang J, Wang S, Zheng W, Cai Y, Wei W, Gordy JT, Markham R, Kong W, Zhang W, Yu XF. Disruption of MDA5-mediated innate immune responses by the 3C proteins of coxsackievirus A16, coxsackievirus A6, and enterovirus D68. J Virol. 2017;91(13).

  181. Xiao H, Li J, Yang X, Li Z, Wang Y, Rui Y, Liu B, Zhang W. Ectopic expression of TRIM25 restores RIG-I expression and IFN production reduced by multiple enteroviruses 3C(pro). Virol Sin. 2021;36(6):1363–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Wang W, Xiao F, Wan P, Pan P, Zhang Y, Liu F, Wu K, Liu Y, Wu J. EV71 3D protein binds with NLRP3 and enhances the assembly of inflammasome complex. PLoS Pathog. 2017;13(1): e1006123.

    Article  PubMed  PubMed Central  Google Scholar 

  183. Koestner W, Spanier J, Klause T, Tegtmeyer PK, Becker J, Herder V, Borst K, Todt D, Lienenklaus S, Gerhauser I, Detje CN, Geffers R, Langereis MA, Vondran FWR, Yuan Q, van Kuppeveld FJM, Ott M, Staeheli P, Steinmann E, Baumgartner W, Wacker F, Kalinke U. Interferon-beta expression and type I interferon receptor signaling of hepatocytes prevent hepatic necrosis and virus dissemination in Coxsackievirus B3-infected mice. PLoS Pathog. 2018;14(8): e1007235.

    Article  PubMed  PubMed Central  Google Scholar 

  184. Han Y, Wang L, Cui J, Song Y, Luo Z, Chen J, Xiong Y, Zhang Q, Liu F, Ho W, Liu Y, Wu K, Wu J. SIRT1 inhibits EV71 genome replication and RNA translation by interfering with the viral polymerase and 5’UTR RNA. J Cell Sci. 2016;129(24):4534–47.

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Wang C, Sun M, Yuan X, Ji L, Jin Y, Cardona CJ, Xing Z. Enterovirus 71 suppresses interferon responses by blocking Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling through inducing karyopherin-alpha1 degradation. J Biol Chem. 2017;292(24):10262–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Chen B, Wang Y, Pei X, Wang S, Zhang H, Peng Y. Cellular caspase-3 contributes to EV-A71 2A(pro)-mediated down-regulation of IFNAR1 at the translation level. Virol Sin. 2020;35(1):64–72.

    Article  CAS  PubMed  Google Scholar 

  187. Wang H, Yuan M, Wang S, Zhang L, Zhang R, Zou X, Wang X, Chen D, Wu Z. STAT3 regulates the type I IFN-mediated antiviral response by interfering with the nuclear entry of STAT1. Int J Mol Sci. 2019;20(19):4870.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Sun M, Lin Q, Wang C, Xing J, Yan K, Liu Z, Jin Y, Cardona CJ, Xing Z. Enterovirus A71 2B inhibits interferon-activated JAK/STAT signaling by inducing caspase-3-dependent karyopherin-alpha1 degradation. Front Microbiol. 2021;12: 762869.

    Article  PubMed  PubMed Central  Google Scholar 

  189. Gu Z, Shi W, Zhang L, Hu Z, Xu C. USP19 suppresses cellular type I interferon signaling by targeting TRAF3 for deubiquitination. Future Microbiol. 2017;12:767–79.

    Article  CAS  PubMed  Google Scholar 

  190. Wang T, Wang B, Huang H, Zhang C, Zhu Y, Pei B, Cheng C, Sun L, Wang J, Jin Q, Zhao Z. Enterovirus 71 protease 2Apro and 3Cpro differentially inhibit the cellular endoplasmic reticulum-associated degradation (ERAD) pathway via distinct mechanisms, and enterovirus 71 hijacks ERAD component p97 to promote its replication. PLoS Pathog. 2017;13(10): e1006674.

    Article  PubMed  PubMed Central  Google Scholar 

  191. You L, Chen J, Liu W, Xiang Q, Luo Z, Wang W, Xu W, Wu K, Zhang Q, Liu Y, Wu J. Enterovirus 71 induces neural cell apoptosis and autophagy through promoting ACOX1 downregulation and ROS generation. Virulence. 2020;11(1):537–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Bonilla FA, Oettgen HC. Adaptive immunity. J Allergy Clin Immunol. 2010;125(2 Suppl 2):S33-40.

    Article  PubMed  Google Scholar 

  193. Nguyet LA, Thanh TT, Nhan LNT, Hong NTT, Nhu LNT, Van HMT, Ny NTH, Anh NT, Han DDK, Tuan HM, Huy VQ, Viet HL, Cuong HQ, Thao NTT, Viet DC, Khanh TH, Thwaites L, Clapham H, Hung NT, Chau NVV, Thwaites G, Ha DQ, van Doorn HR, Tan LV. Neutralizing antibodies against enteroviruses in patients with hand, foot and mouth disease. Emerg Infect Dis. 2020;26(2):298–306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Yang C, Deng C, Wan J, Zhu L, Leng Q. Neutralizing antibody response in the patients with hand, foot and mouth disease to enterovirus 71 and its clinical implications. Virol J. 2011;8:306.

    Article  PubMed  PubMed Central  Google Scholar 

  195. Qiu Q, Zhou J, Cheng Y, Zhou Y, Liang L, Cui P, Xue Y, Wang L, Wang K, Wang H, Li P, Chen J, Li Y, Turtle L, Yu H. Kinetics of the neutralising antibody response in patients with hand, foot, and mouth disease caused by EV-A71: a longitudinal cohort study in Zhengzhou during 2017–2019. EBioMedicine. 2021;68: 103398.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Fang Y, Lian C, Huang D, Xu L. Analysis of clinical related factors of neonatal hand-foot-mouth disease complicated with encephalitis. Front Neurol. 2020;11: 543013.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Chang LY, Hsiung CA, Lu CY, Lin TY, Huang FY, Lai YH, Chiang YP, Chiang BL, Lee CY, Huang LM. Status of cellular rather than humoral immunity is correlated with clinical outcome of enterovirus 71. Pediatr Res. 2006;60(4):466–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Lin YW, Chang KC, Kao CM, Chang SP, Tung YY, Chen SH. Lymphocyte and antibody responses reduce enterovirus 71 lethality in mice by decreasing tissue viral loads. J Virol. 2009;83(13):6477–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Zhao MQ, Wang LH, Lian GW, Lin ZF, Li YH, Guo M, Chen Y, Liu XM, Zhu B. Characterization of lymphocyte subsets in peripheral blood cells of children with EV71 infection. J Microbiol Immunol Infect. 2020;53(5):705–14.

    Article  CAS  PubMed  Google Scholar 

  200. Li S, Cai C, Feng J, Li X, Wang Y, Yang J, Chen Z. Peripheral T lymphocyte subset imbalances in children with enterovirus 71-induced hand, foot and mouth disease. Virus Res. 2014;180:84–91.

    Article  CAS  PubMed  Google Scholar 

  201. Li Q, Wang Y, Bian Z, Gao Y, Zeng Y, Tang L, Tang T, Tian Y, Guo W. Abnormalities of ILC1 in children with hand, foot and mouth disease during enterovirus 71 infection. Virology. 2020;551:36–45.

    Article  CAS  PubMed  Google Scholar 

  202. Zhang W, Huang Z, Huang M, Zeng J. Predicting severe enterovirus 71-infected hand, foot, and mouth disease: cytokines and chemokines. Mediators Inflamm. 2020;2020:9273241.

    Article  PubMed  PubMed Central  Google Scholar 

  203. Zhang Y, Liu H, Wang L, Yang F, Hu Y, Ren X, Li G, Yang Y, Sun S, Li Y, Chen X, Li X, Jin Q. Comparative study of the cytokine/chemokine response in children with differing disease severity in enterovirus 71-induced hand, foot, and mouth disease. PLoS ONE. 2013;8(6): e67430.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Duan G, Yang H, Shi L, Sun W, Sui M, Zhang R, Wang X, Wang F, Zhang W, Xi Y, Fan Q. Serum inflammatory cytokine levels correlate with hand-foot-mouth disease severity: a nested serial case-control study. PLoS ONE. 2014;9(11): e112676.

    Article  PubMed  PubMed Central  Google Scholar 

  205. Avau A, Mitera T, Put S, Put K, Brisse E, Filtjens J, Uyttenhove C, Van Snick J, Liston A, Leclercq G, Billiau AD, Wouters CH, Matthys P. Systemic juvenile idiopathic arthritis-like syndrome in mice following stimulation of the immune system with Freund’s complete adjuvant: regulation by interferon-gamma. Arthritis Rheumatol. 2014;66(5):1340–51.

    Article  CAS  PubMed  Google Scholar 

  206. Fajgenbaum DC, June CH. Cytokine Storm. N Engl J Med. 2020;383(23):2255–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  207. Lin TY, Hsia SH, Huang YC, Wu CT, Chang LY. Proinflammatory cytokine reactions in enterovirus 71 infections of the central nervous system. Clin Infect Dis. 2003;36(3):269–74.

    Article  CAS  PubMed  Google Scholar 

  208. Wang SM, Lei HY, Yu CK, Wang JR, Su IJ, Liu CC. Acute chemokine response in the blood and cerebrospinal fluid of children with enterovirus 71-associated brainstem encephalitis. J Infect Dis. 2008;198(7):1002–6.

    Article  CAS  PubMed  Google Scholar 

  209. Shen FH, Tsai CC, Wang LC, Chang KC, Tung YY, Su IJ, Chen SH. Enterovirus 71 infection increases expression of interferon-gamma-inducible protein 10 which protects mice by reducing viral burden in multiple tissues. J Gen Virol. 2013;94(Pt 5):1019–27.

    Article  CAS  PubMed  Google Scholar 

  210. Sun Z, Li W, Xu J, Ren K, Gao F, Jiang Z, Ji F, Pan D. Proteomic analysis of cerebrospinal fluid in children with acute enterovirus-associated meningoencephalitis identifies dysregulated host processes and potential biomarkers. J Proteome Res. 2020;19(8):3487–98.

    Article  CAS  PubMed  Google Scholar 

  211. Liang R, Chen S, Jin Y, Tao L, Ji W, Zhu P, Li D, Zhang Y, Zhang W, Duan G. The CXCL10/CXCR3 axis promotes disease pathogenesis in mice upon CVA2 infection. Microbiol Spectr. 2022;10(3): e0230721.

    Article  PubMed  Google Scholar 

  212. Wang SM, Lei HY, Huang KJ, Wu JM, Wang JR, Yu CK, Su IJ, Liu CC. Pathogenesis of enterovirus 71 brainstem encephalitis in pediatric patients: roles of cytokines and cellular immune activation in patients with pulmonary edema. J Infect Dis. 2003;188(4):564–70.

    Article  CAS  PubMed  Google Scholar 

  213. Li ML, Hsu TA, Chen TC, Chang SC, Lee JC, Chen CC, Stollar V, Shih SR. The 3C protease activity of enterovirus 71 induces human neural cell apoptosis. Virology. 2002;293(2):386–95.

    Article  CAS  PubMed  Google Scholar 

  214. Li H, Bai Z, Li C, Sheng C, Zhao X. EV71 infection induces cell apoptosis through ROS generation and SIRT1 activation. J Cell Biochem. 2020;121(10):4321–31.

    Article  CAS  PubMed  Google Scholar 

  215. Koyuncu OO, Hogue IB, Enquist LW. Virus infections in the nervous system. Cell Host Microbe. 2013;13(4):379–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Lai RH, Chow YH, Chung NH, Chen TC, Shie FS, Juang JL. Neurotropic EV71 causes encephalitis by engaging intracellular TLR9 to elicit neurotoxic IL12-p40-iNOS signaling. Cell Death Dis. 2022;13(4):328.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Barral PM, Sarkar D, Fisher PB, Racaniello VR. RIG-I is cleaved during picornavirus infection. Virology. 2009;391(2):171–6.

    Article  CAS  PubMed  Google Scholar 

  218. Chang Z, Wang Y, Bian L, Liu Q, Long JE. Enterovirus 71 antagonizes the antiviral activity of host STAT3 and IL-6R with partial dependence on virus-induced miR-124. J Gen Virol. 2017;98(12):3008–25.

    Article  CAS  PubMed  Google Scholar 

  219. Chang LY, Lin TY, Hsu KH, Huang YC, Lin KL, Hsueh C, Shih SR, Ning HC, Hwang MS, Wang HS, Lee CY. Clinical features and risk factors of pulmonary oedema after enterovirus-71-related hand, foot, and mouth disease. Lancet. 1999;354(9191):1682–6.

    Article  CAS  PubMed  Google Scholar 

  220. Chang LY, Huang YC, Lin TY. Fulminant neurogenic pulmonary oedema with hand, foot, and mouth disease. Lancet. 1998;352(9125):367–8.

    Article  CAS  PubMed  Google Scholar 

  221. Yan JJ, Wang JR, Liu CC, Yang HB, Su IJ. An outbreak of enterovirus 71 infection in Taiwan 1998: a comprehensive pathological, virological, and molecular study on a case of fulminant encephalitis. J Clin Virol. 2000;17(1):13–22.

    Article  CAS  PubMed  Google Scholar 

  222. Hsueh C, Jung SM, Shih SR, Kuo TT, Shieh WJ, Zaki S, Lin TY, Chang LY, Ning HC, Yen DC. Acute encephalomyelitis during an outbreak of enterovirus type 71 infection in Taiwan: report of an autopsy case with pathologic, immunofluorescence, and molecular studies. Mod Pathol. 2000;13(11):1200–5.

    Article  CAS  PubMed  Google Scholar 

  223. Wang SM, Liu CC, Tseng HW, Wang JR, Huang CC, Chen YJ, Yang YJ, Lin SJ, Yeh TF. Clinical spectrum of enterovirus 71 infection in children in southern Taiwan, with an emphasis on neurological complications. Clin Infect Dis. 1999;29(1):184–90.

    Article  CAS  PubMed  Google Scholar 

  224. Hsu YH, Kao SJ, Lee RP, Chen HI. Acute pulmonary oedema: rare causes and possible mechanisms. Clin Sci (Lond). 2003;104(3):259–64.

    Article  CAS  PubMed  Google Scholar 

  225. Wu JM, Wang JN, Tsai YC, Liu CC, Huang CC, Chen YJ, Yeh TF. Cardiopulmonary manifestations of fulminant enterovirus 71 infection. Pediatrics. 2002;109(2):E26.

    Article  PubMed  Google Scholar 

  226. Jin Y, Sun T, Zhou G, Li D, Chen S, Zhang W, Li X, Zhang R, Yang H, Duan G. Pathogenesis study of enterovirus 71 using a novel human SCARB2 knock-in mouse model. mSphere. 2021;6(2):e01048-e1120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Li D, Sun T, Tao L, Ji W, Zhu P, Liang R, Zhang Y, Chen S, Yang H, Jin Y, Duan G. A mouse-adapted CVA6 strain exhibits neurotropism and triggers systemic manifestations in a novel murine model. Emerg Microbes Infect. 2022;11(1):2248–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Ji W, Hu Q, Zhang M, Zhang C, Chen C, Yan Y, Zhang X, Chen S, Tao L, Zhang W, Jin Y, Duan G. The disruption of the endothelial barrier contributes to acute lung injury induced by coxsackievirus A2 infection in mice. Int J Mol Sci. 2021;22(18):9895.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  229. Lum LC, Wong KT, Lam SK, Chua KB, Goh AY, Lim WL, Ong BB, Paul G, AbuBakar S, Lambert M. Fatal enterovirus 71 encephalomyelitis. J Pediatr. 1998;133(6):795–8.

    Article  CAS  PubMed  Google Scholar 

  230. Huang FL, Jan SL, Chen PY, Chi CS, Wang TM, Fu YC, Tsai CR, Chang Y. Left ventricular dysfunction in children with fulminant enterovirus 71 infection: an evaluation of the clinical course. Clin Infect Dis. 2002;34(7):1020–4.

    Article  PubMed  Google Scholar 

  231. Fu YC, Chi CS, Chiu YT, Hsu SL, Hwang B, Jan SL, Chen PY, Huang FL, Chang Y. Cardiac complications of enterovirus rhombencephalitis. Arch Dis Child. 2004;89(4):368–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Jan SL, Lin MC, Chan SC, Lee HF, Chen PY, Huang FL. Urine catecholamines in children with severe Enterovirus A71 infection: comparison with paediatric septic shock. Biomarkers. 2019;24(3):277–85.

    Article  CAS  PubMed  Google Scholar 

  233. Fu X, Mao L, Wan Z, Xu R, Ma Y, Shen L, Jin X, Zhang C. High proportion of coxsackievirus B3 genotype A in hand, foot and mouth disease in Zhenjiang, China, 2011–2016. Int J Infect Dis. 2019;87:1–7.

    Article  CAS  PubMed  Google Scholar 

  234. Lasrado N, Reddy J. An overview of the immune mechanisms of viral myocarditis. Rev Med Virol. 2020;30(6):1–14.

    Article  CAS  PubMed  Google Scholar 

  235. Molet L, Saloum K, Marque-Juillet S, Garbarg-Chenon A, Henquell C, Schuffenecker I, Peigue-Lafeuille H, Rozenberg F, Mirand A. Enterovirus infections in hospitals of Ile de France region over 2013. J Clin Virol. 2016;74:37–42.

    Article  PubMed  Google Scholar 

  236. Ji W, Qin L, Tao L, Zhu P, Liang R, Zhou G, Chen S, Zhang W, Yang H, Duan G, Jin Y. Neonatal murine model of coxsackievirus A2 infection for the evaluation of antiviral therapeutics and vaccination. Front Microbiol. 2021;12: 658093.

    Article  PubMed  PubMed Central  Google Scholar 

  237. Huang X, Zhang X, Wang F, Wei H, Ma H, Sui M, Lu J, Wang H, Dumler JS, Sheng G, Xu B. Clinical efficacy of therapy with recombinant human interferon alpha1b in hand, foot, and mouth disease with enterovirus 71 infection. PLoS ONE. 2016;11(2): e0148907.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Lin H, Huang L, Zhou J, Lin K, Wang H, Xue X, Xia C. Efficacy and safety of interferon-alpha2b spray in the treatment of hand, foot, and mouth disease: a multicenter, randomized, double-blind trial. Adv Virol. 2016;161(11):3073–80.

    CAS  Google Scholar 

  239. Abzug MJ. The enteroviruses: problems in need of treatments. J Infect. 2014;68(Suppl 1):S108-114.

    Article  PubMed  Google Scholar 

  240. Lin JY, Kung YA, Shih SR. Antivirals and vaccines for Enterovirus A71. J Biomed Sci. 2019;26(1):65.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Lin TY, Chang LY, Hsia SH, Huang YC, Chiu CH, Hsueh C, Shih SR, Liu CC, Wu MH. The 1998 enterovirus 71 outbreak in Taiwan: pathogenesis and management. Clin Infect Dis. 2002;34(Suppl 2):S52-57.

    Article  PubMed  Google Scholar 

  242. Wang SM, Lei HY, Huang MC, Su LY, Lin HC, Yu CK, Wang JL, Liu CC. Modulation of cytokine production by intravenous immunoglobulin in patients with enterovirus 71-associated brainstem encephalitis. J Clin Virol. 2006;37(1):47–52.

    Article  CAS  PubMed  Google Scholar 

  243. Ooi MH, Wong SC, Podin Y, Akin W, del Sel S, Mohan A, Chieng CH, Perera D, Clear D, Wong D, Blake E, Cardosa J, Solomon T. Human enterovirus 71 disease in Sarawak, Malaysia: a prospective clinical, virological, and molecular epidemiological study. Clin Infect Dis. 2007;44(5):646–56.

    Article  PubMed  Google Scholar 

  244. Cai K, Wang Y, Guo Z, Yu H, Li H, Zhang L, Xu S, Zhang Q. Clinical characteristics and managements of severe hand, foot and mouth disease caused by enterovirus A71 and coxsackievirus A16 in Shanghai, China. BMC Infect Dis. 2019;19(1):285.

    Article  PubMed  PubMed Central  Google Scholar 

  245. Liu J, Qi J. Prevalence and management of severe hand, foot, and mouth disease in Xiangyang, China from 2008–2013. J Med Virol. 2020;33:340.

    Google Scholar 

  246. Jiao W, Tan SR, Huang YF, Mu LH, Yang Y, Wang Y, Wu XE. The effectiveness of different doses of intravenous immunoglobulin on severe hand, foot and mouth disease: a meta-analysis. Med Princ Pract. 2019;28(3):256–63.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Wang SM, Liu CC. Enterovirus 71: epidemiology, pathogenesis and management. Expert Rev Anti Infect Ther. 2009;7(6):735–42.

    Article  PubMed  Google Scholar 

  248. Yan Z, Shang Y, Li F, Xie F, Qian H, Zhang Y, Yue B. Therapeutic efficacy of phentolamine in the management of severe hand, foot and mouth disease combined with pulmonary edema. Exp Ther Med. 2017;13(4):1403–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Wang SM. Milrinone in enterovirus 71 brain stem encephalitis. Front Pharmacol. 2016;7:82.

    Article  PubMed  PubMed Central  Google Scholar 

  250. Yang TT, Huang LM, Lu CY, Kao CL, Lee WT, Lee PI, Chen CM, Huang FY, Lee CY, Chang LY. Clinical features and factors of unfavorable outcomes for non-polio enterovirus infection of the central nervous system in northern Taiwan, 1994–2003. J Microbiol Immunol Infect. 2005;38(6):417–24.

    PubMed  Google Scholar 

  251. Li XH, Li SJ, Xu Y, Wei D, Shi QS, Zhu QX, Yang T, Ding JB, Tian YM, Huang JH, Wang K, Wen T, Zhang X. Effect of integrated Chinese and Western medicine therapy on severe hand, foot and mouth disease: a prospective, randomized, controlled trial. Chin J Integr Med. 2017;23(12):887–92.

    Article  PubMed  Google Scholar 

  252. Li X, Zhang C, Shi Q, Yang T, Zhu Q, Tian Y, Lu C, Zhang Z, Jiang Z, Zhou H, Wen X, Yang H, Ding X, Liang L, Liu Y, Wang Y, Lu A. Improving the efficacy of conventional therapy by adding andrographolide sulfonate in the treatment of severe hand, foot, and mouth disease: a randomized controlled trial. Evid Based Complement Alternat Med. 2013;2013: 316250.

    PubMed  PubMed Central  Google Scholar 

  253. Lin H, Zhou J, Lin K, Wang H, Liang Z, Ren X, Huang L, Xia C. Efficacy of Scutellaria baicalensis for the treatment of hand, foot, and mouth disease associated with encephalitis in patients infected with EV71: a multicenter, retrospective analysis. Biomed Res Int. 2016;2016:5697571.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Wang C, Cui Y, Zhu Y, Wang F, Rong Q, Zhang Y. Continuous hemodiafiltration as a rescue therapy for patients with cardiopulmonary failure caused by enterovirus-71: a retrospective observational study in a PICU. BMC Infect Dis. 2019;19(1):866.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Jones E, Pillay TD, Liu F, Luo L, Bazo-Alvarez JC, Yuan C, Zhao S, Chen Q, Li Y, Liao Q, Yu H, Rogier van Doorn H, Sabanathan S. Outcomes following severe hand foot and mouth disease: a systematic review and meta-analysis. Eur J Paediatr Neurol. 2018;22(5):763–73.

    Article  PubMed  PubMed Central  Google Scholar 

  256. Ji H, Fan H, Ai J, Shi C, Bi J, Chen YH, Lu XP, Chen QH, Tian JM, Bao CJ, Zhang XF, Jin Y. Neurocognitive deficits and sequelae following severe hand, foot, and mouth disease from 2009 to 2017, in JiangSu Province, China: a long-term follow-up study. Int J Infect Dis. 2022;115:245–55.

    Article  PubMed  Google Scholar 

  257. Prager P, Nolan M, Andrews IP, Williams GD. Neurogenic pulmonary edema in enterovirus 71 encephalitis is not uniformly fatal but causes severe morbidity in survivors. Pediatr Crit Care Med. 2003;4(3):377–81.

    Article  PubMed  Google Scholar 

  258. Chang LY, Huang LM, Gau SS, Wu YY, Hsia SH, Fan TY, Lin KL, Huang YC, Lu CY, Lin TY. Neurodevelopment and cognition in children after enterovirus 71 infection. N Engl J Med. 2007;356(12):1226–34.

    Article  CAS  PubMed  Google Scholar 

  259. Tsou YA, Cheng YK, Chung HK, Yeh YC, Lin CD, Tsai MH, Chang JS. Upper aerodigestive tract sequelae in severe enterovirus 71 infection: predictors and outcome. Int J Pediatr Otorhinolaryngol. 2008;72(1):41–7.

    Article  PubMed  Google Scholar 

  260. Liang L, Cheng Y, Li Y, Shang Q, Huang J, Ma C, Fang S, Long L, Zhou C, Chen Z, Cui P, Lv N, Lou P, Cui Y, Sabanathan S, van Doorn HR, Luan R, Turtle L, Yu H. Long-term neurodevelopment outcomes of hand, foot and mouth disease inpatients infected with EV-A71 or CV-A16, a retrospective cohort study. Emerg Microbes Infect. 2021;10(1):545–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  261. Dalman C, Allebeck P, Gunnell D, Harrison G, Kristensson K, Lewis G, Lofving S, Rasmussen F, Wicks S, Karlsson H. Infections in the CNS during childhood and the risk of subsequent psychotic illness: a cohort study of more than one million Swedish subjects. Am J Psychiatry. 2008;165(1):59–65.

    Article  PubMed  Google Scholar 

  262. Gau SS, Chang LY, Huang LM, Fan TY, Wu YY, Lin TY. Attention-deficit/hyperactivity-related symptoms among children with enterovirus 71 infection of the central nervous system. Pediatrics. 2008;122(2):e452-458.

    Article  PubMed  Google Scholar 

  263. Pedersen EMJ, Kohler-Forsberg O, Nordentoft M, Christensen RHB, Mortensen PB, Petersen L, Benros ME. Infections of the central nervous system as a risk factor for mental disorders and cognitive impairment: a nationwide register-based study. Brain Behav Immun. 2020;88:668–74.

    Article  PubMed  Google Scholar 

  264. Teoh HL, Mohammad SS, Britton PN, Kandula T, Lorentzos MS, Booy R, Jones CA, Rawlinson W, Ramachandran V, Rodriguez ML, Andrews PI, Dale RC, Farrar MA, Sampaio H. Clinical characteristics and functional motor outcomes of enterovirus 71 neurological disease in children. JAMA Neurol. 2016;73(3):300–7.

    Article  PubMed  Google Scholar 

  265. Lian ZY, Li HH, Zhang B, Dong YH, Deng WX, Liu J, Luo XN, Huang B, Liang CH, Zhang SX. Neuro-magnetic resonance imaging in hand, foot, and mouth disease: finding in 412 patients and prognostic features. J Comput Assist Tomogr. 2017;41(6):861–7.

    Article  PubMed  PubMed Central  Google Scholar 

  266. Kim YJ, Kim TG. Pseudomembranous conjunctivitis with hand, foot and mouth disease in a pregnant woman: a case report. BMC Ophthalmol. 2021;21(1):113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  267. Haamann P, Kessel L, Larsen M. Monofocal outer retinitis associated with hand, foot, and mouth disease caused by coxsackievirus. Am J Ophthalmol. 2000;129(4):552–3.

    Article  CAS  PubMed  Google Scholar 

  268. Ng SK, Ebneter A, Gilhotra JS. Atypical findings in delayed presentation of unilateral acute idiopathic maculopathy. Int Ophthalmol. 2013;33(4):387–9.

    Article  PubMed  Google Scholar 

  269. Wei SH, Huang YP, Liu MC, Tsou TP, Lin HC, Lin TL, Tsai CY, Chao YN, Chang LY, Hsu CM. An outbreak of coxsackievirus A6 hand, foot, and mouth disease associated with onychomadesis in Taiwan, 2010. BMC Infect Dis. 2011;11:346.

    Article  PubMed  PubMed Central  Google Scholar 

  270. Chiu HH, Liu MT, Chung WH, Ko YS, Lu CF, Lan CE, Lu CW, Wei KC. The mechanism of onychomadesis (nail shedding) and beau’s lines following hand-foot-mouth disease. Viruses. 2019;11(6):522.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Akpolat ND, Karaca N. Nail changes secondary to hand-foot-mouth disease. Turk J Pediatr. 2016;58(3):287–90.

    Article  PubMed  Google Scholar 

  272. Long DL, Zhu SY, Li CZ, Chen CY, Du WT, Wang X. Late-onset nail changes associated with hand, foot, and mouth disease: a clinical analysis of 56 cases. Pediatr Dermatol. 2016;33(4):424–8.

    Article  PubMed  Google Scholar 

  273. Abramovici G, Keoprasom N, Winslow CY, Tosti A. Onycholysis and subungual haemorrhages in a patient with hand, foot and mouth disease. Br J Dermatol. 2014;170(3):748–9.

    Article  CAS  PubMed  Google Scholar 

  274. Gan XL, Zhang TD. Onychomadesis after hand-foot-and-mouth disease. CMAJ. 2017;189(7):E279.

    Article  PubMed  PubMed Central  Google Scholar 

  275. Tan ZH, Koh MJ. Nail shedding following hand, foot and mouth disease. Arch Dis Child. 2013;98(9):665.

    Article  PubMed  Google Scholar 

  276. Yuksel S, Evrengul H, Ozhan B, Yuksel G. Onychomadesis-a late complication of hand-foot-mouth disease. J Pediatr. 2016;174:274.

    Article  PubMed  Google Scholar 

  277. Lee ZM, Huang YH, Ho SC, Kuo HC. Correlation of symptomatic enterovirus infection and later risk of allergic diseases via a population-based cohort study. Medicine (Baltimore). 2017;96(4): e5827.

    Article  PubMed  Google Scholar 

  278. Yeh JJ, Lin CL, Hsu WH. Effect of enterovirus infections on asthma in young children: a national cohort study. Eur J Clin Invest. 2017;47(12):e12844.

    Article  Google Scholar 

  279. Broccolo F, Drago F, Ciccarese G, Genoni A, Porro A, Parodi A, Chumakov K, Toniolo A. Possible long-term sequelae in hand, foot, and mouth disease caused by Coxsackievirus A6. J Am Acad Dermatol. 2019;80(3):804–6.

    Article  PubMed  Google Scholar 

  280. Weng KP, Cheng-Chung Wei J, Hung YM, Huang SH, Chien KJ, Lin CC, Huang SM, Lin CL, Cheng MF. Enterovirus infection and subsequent risk of Kawasaki disease: a population-based cohort study. Pediatric Infect Dis J. 2018;37(4):310–5.

    Article  Google Scholar 

  281. Lin JN, Lin CL, Yang CH, Lin MC, Lai CH, Lin HH, Kao CH. Risk of nephrotic syndrome following enteroviral infection in children: a nationwide retrospective cohort study. PLoS ONE. 2016;11(8): e0161004.

    Article  PubMed  PubMed Central  Google Scholar 

  282. Lin JN, Lin CL, Lin MC, Lai CH, Lin HH, Yang CH, Sung FC, Kao CH. Risk of leukaemia in children infected with enterovirus: a nationwide, retrospective, population-based, Taiwanese-registry, cohort study. Lancet Oncol. 2015;16(13):1335–43.

    Article  PubMed  Google Scholar 

  283. Foo DG, Alonso S, Phoon MC, Ramachandran NP, Chow VT, Poh CL. Identification of neutralizing linear epitopes from the VP1 capsid protein of Enterovirus 71 using synthetic peptides. Virus Res. 2007;125(1):61–8.

    Article  CAS  PubMed  Google Scholar 

  284. Premanand B, Kiener TK, Meng T, Tan YR, Jia Q, Chow VT, Kwang J. Induction of protective immune responses against EV71 in mice by baculovirus encoding a novel expression cassette for capsid protein VP1. Antiviral Res. 2012;95(3):311–5.

    Article  CAS  PubMed  Google Scholar 

  285. Yang Z, Gao F, Wang X, Shi L, Zhou Z, Jiang Y, Ma X, Zhang C, Zhou C, Zeng X, Liu G, Fan J, Mao Q, Shi L. Development and characterization of an enterovirus 71 (EV71) virus-like particles (VLPs) vaccine produced in Pichia pastoris. Hum Vaccin Immunother. 2020;16(7):1602–10.

    Article  CAS  PubMed  Google Scholar 

  286. Li R, Liu L, Mo Z, Wang X, Xia J, Liang Z, Zhang Y, Li Y, Mao Q, Wang J, Jiang L, Dong C, Che Y, Huang T, Jiang Z, Xie Z, Wang L, Liao Y, Liang Y, Nong Y, Liu J, Zhao H, Na R, Guo L, Pu J, Yang E, Sun L, Cui P, Shi H, Wang J, Li Q. An inactivated enterovirus 71 vaccine in healthy children. N Engl J Med. 2014;370(9):829–37.

    Article  CAS  PubMed  Google Scholar 

  287. Li J, Chang J, Liu X, Yang J, Guo H, Wei W, Zhang W, Yu XF. Protection from lethal challenge in a neonatal mouse model by circulating recombinant form coxsackievirus A16 vaccine candidates. J Gen Virol. 2014;95(Pt 5):1083–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  288. Zhang Z, Dong Z, Li J, Carr MJ, Zhuang D, Wang J, Zhang Y, Ding S, Tong Y, Li D, Shi W. Protective efficacies of formaldehyde-inactivated whole-virus vaccine and antivirals in a murine model of coxsackievirus A10 infection. J Virol. 2017;91(13):e00333-e417.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Jin WP, Lu J, Zhang XY, Wu J, Wei ZN, Mai JY, Qian SS, Yu YT, Meng SL, Wang ZJ, Shen S. Efficacy of coxsackievirus A5 vaccine candidates in an actively immunized mouse model. J Virol. 2021;95(6):e01743-e1820.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  290. Qian SS, Wei ZN, Jin WP, Wu J, Zhou YP, Meng SL, Guo J, Wang ZJ, Shen S. Efficacy of a coxsackievirus A6 vaccine candidate in an actively immunized mouse model. Emerg Microbes Infect. 2021;10(1):763–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Cai Y, Ku Z, Liu Q, Leng Q, Huang Z. A combination vaccine comprising of inactivated enterovirus 71 and coxsackievirus A16 elicits balanced protective immunity against both viruses. Vaccine. 2014;32(21):2406–12.

    Article  CAS  PubMed  Google Scholar 

  292. Fan S, Liao Y, Jiang G, Jiang L, Wang L, Xu X, Feng M, Yang E, Zhang Y, Cui W, Li Q. Study of integrated protective immunity induced in rhesus macaques by the intradermal administration of a bivalent EV71-CA16 inactivated vaccine. Vaccine. 2020;38(8):2034–44.

    Article  CAS  PubMed  Google Scholar 

  293. Zhang Z, Dong Z, Wang Q, Carr MJ, Li J, Liu T, Li D, Shi W. Characterization of an inactivated whole-virus bivalent vaccine that induces balanced protective immunity against coxsackievirus A6 and A10 in mice. Vaccine. 2018;36(46):7095–104.

    Article  CAS  PubMed  Google Scholar 

  294. Caine EA, Fuchs J, Das SC, Partidos CD, Osorio JE. Efficacy of a trivalent hand, foot, and mouth disease vaccine against enterovirus 71 and coxsackieviruses A16 and A6 in mice. Viruses. 2015;7(11):5919–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  295. Lim H, In HJ, Lee JA, Sik Yoo J, Lee SW, Chung GT, Choi YK, Chung JK, Cho SJ, Lee JW. The immunogenicity and protection effect of an inactivated coxsackievirus A6, A10, and A16 vaccine against hand, foot, and mouth disease. Vaccine. 2018;36(24):3445–52.

    Article  CAS  PubMed  Google Scholar 

  296. Zhang W, Dai W, Zhang C, Zhou Y, Xiong P, Wang S, Ye X, Liu Q, Zhou D, Huang Z. A virus-like particle-based tetravalent vaccine for hand, foot, and mouth disease elicits broad and balanced protective immunity. Emerg Microbes Infect. 2018;7(1):94.

    Article  PubMed  PubMed Central  Google Scholar 

  297. He X, Zhang M, Zhao C, Zheng P, Zhang X, Xu J. From monovalent to multivalent vaccines, the exploration for potential preventive strategies against hand, foot, and mouth disease (HFMD). Virol Sin. 2021;36(2):167–75.

    Article  PubMed  Google Scholar 

  298. Anasir MI, Poh CL. Advances in antigenic peptide-based vaccine and neutralizing antibodies against viruses causing hand, foot, and mouth disease. Int J Mol Sci. 2019;20(6):1256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  299. Fang CY, Liu CC. Novel strategies for the development of hand, foot, and mouth disease vaccines and antiviral therapies. Expert Opin Drug Discov. 2022;17(1):27–39.

    Article  CAS  PubMed  Google Scholar 

  300. Abedi GR, Watson JT, Nix WA, Oberste MS, Gerber SI. Enterovirus and parechovirus surveillance—United States, 2014–2016. MMWR Morb Mortal Wkly Rep. 2018;67(18):515–8.

    Article  PubMed  PubMed Central  Google Scholar 

  301. Chiu ML, Luo ST, Chen YY, Chung WY, Duong V, Dussart P, Chan YF, Perera D, Ooi MH, Thao NTT, Truong HK, Lee MS. Establishment of Asia-Pacific Network for Enterovirus Surveillance. Vaccine. 2020;38(1):1–9.

    Article  PubMed  Google Scholar 

  302. Harvala H, Benschop KSM, Berginc N, Midgley S, Wolthers K, Simmonds P, Feeney S, Bailly JL, Mirand A, Fischer TK, and On Behalf Of The Enpen Hospital-Based Surveillance N. European Non-Polio Enterovirus Network: introduction of Hospital-Based Surveillance Network to Understand the True Disease Burden of Non-Polio Enterovirus and Parechovirus Infections in Europe. Microorganisms. 1827;9(9):2021.

    Google Scholar 

  303. Cheng Q, Collender PA, Heaney AK, McLoughlin A, Yang Y, Zhang Y, Head JR, Dasan R, Liang S, Lv Q, Liu Y, Yang C, Chang HH, Waller LA, Zelner J, Lewnard JA, Remais JV. Optimizing laboratory-based surveillance networks for monitoring multi-genotype or multi-serotype infections. PLoS Comput Biol. 2022;18(9): e1010575.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  304. Yao X, Bian LL, Lu WW, Li JX, Mao QY, Wang YP, Gao F, Wu X, Ye Q, Xu M, Li XL, Zhu FC, Liang ZL. Enterovirus spectrum from the active surveillance of hand foot and mouth disease patients under the clinical trial of inactivated Enterovirus A71 vaccine in Jiangsu, China, 2012–2013. J Med Virol. 2015;87(12):2009–17.

    Article  CAS  PubMed  Google Scholar 

  305. Zhang X, Zhang Y, Li H, Liu L. Hand-foot-and-mouth disease-associated enterovirus and the development of multivalent HFMD vaccines. Int J Mol Sci. 2022;24(1).

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (NO.82273695, NO.82002147 and NO.82073618); China Postdoctoral Science Foundation (NO.2019M662543); Key Scientific Research Project of Henan Institution of Higher Education (NO.21A310026). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, YJ and GD; Data curation, PZ and SC; Formal analysis, PZ, DL and ZL; Funding acquisition, SC and YJ; Investigation, WJ, DL, PZ, CY, BD, SH and ZL; Methodology, PZ, WJ and YJ; Project administration, PZ, WJ, YJ and GD; Resources, WJ and PZ; Software, PZ, DL and ZL; Supervision, GD; Validation, ZL and DL; Visualization, WJ and SC; Writing—original draft, PZ and YJ; Writing—review and editing, PZ, WJ, YJ and GD All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Yuefei Jin or Guangcai Duan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have declared that no competing interests exist.

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 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhu, P., Ji, W., Li, D. et al. Current status of hand-foot-and-mouth disease. J Biomed Sci 30, 15 (2023). https://doi.org/10.1186/s12929-023-00908-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-023-00908-4

Keywords