2022 Article 775
2022 Article 775
2022 Article 775
Monkeypox is a zoonotic viral disease caused by the rates in patients vaccinated against smallpox4. Recently,
monkeypox virus (MPXV), an orthopoxvirus belong- a study on 528 infections diagnosed during this current
ing to the Poxviridae family of viruses1. MPXV was outbreak reported that only 9% of the individuals who
first identified in 1958 in research monkeys that were were infected received prior smallpox vaccination8.
shipped from Singapore2, which is the likely reason for Interestingly, one characteristic of the recent outbreaks
the disease being called ‘monkeypox’. However, the nat- is a disproportionate number of infections in men who
ural hosts of MPXV are more likely to be rodents and have sex with men (MSM)8,9.
other small mammals3. The Orthopoxvirus genus also Phylogenetically, MPXV can be divided into two dis-
includes variola virus (VARV), the causative agent of tinct clades — Central African (also commonly known
lethal smallpox disease. The symptoms of monkeypox as Congo Basin) and West African. Depending on the
in humans are relatively similar to those of smallpox, but source of the West African MPXV, sequence similarity of
with a lower mortality rate1,4. ~95%10 or >99% between the two clades11 was reported.
Sporadic cases of MPXV in humans were first iden- The Central African clade is generally considered to be
tified in the 1970s in several African countries, but the more virulent, with an average fatality rate of 10.6% (95%
virus became more widespread within the African con- CI 8.4–13.3%) compared with the 3.6% (95% CI 1.7–6.8%)
tinent over the past 20 years (see Box 1 for details). Since reported for the West African clade12. MPXV of the
May 2022, there has been a drastic increase in the num- Central African clade has been identified in cases appear-
ber of MPXV cases worldwide (Fig. 1), leading the World ing in the Democratic Republic of the Congo (DRC) and
Health Organization (WHO) to declare the monkeypox South Sudan12, whereas cases in Nigeria between 2010
outbreak a global health emergency5. and 2019 appeared to be due to the West African clade12.
Possible reasons for this current outbreak could be Cases reported outside Africa, including those currently
attributed to waning smallpox immunity in the general in circulation, have all been caused by the West African
population and termination of the smallpox vaccination clade9,13. Whether genetic changes in the MPXV genome
regime6. Vaccination against smallpox has been shown could be responsible for this current outbreak is currently
to offer protection against monkeypox. An early study being investigated14 (see Box 2 for details).
from Zaire in 1988 (ref.7) reported that individuals vac- Here, we discuss our current understanding of the
cinated against smallpox (during a national smallpox transmission and immunopathogenesis of MPXV and
vaccination campaign beginning 12 years before the the unique epidemiological and pathological character-
start of data collection) were approximately 85% less istics observed in this outbreak. Specifically, we exam-
✉e-mail: Lisa_Ng@ likely to contract monkeypox than those who were not ine the potential mechanisms of host immunity against
idlabs.a-star.edu.sg vaccinated. In another study, severe complications and MPXV, drawing parallels from other poxviruses where
https://doi.org/10.1038/ long-term effects of MPXV infection were found to necessary. We also discuss vaccines and therapeutics,
s41577-022-00775-4 be less common (39.5% versus 74%) with lower death and highlight remaining critical gaps in our knowledge.
0123456789();:
Reviews
Of note, nomenclature for- orthopoxviruses gene/pro- report from Portugal suggested that inguinal lymphad-
tein orthologues is very complex. In this Review, we enopathy was more common than cervical and axillary
refer to orthopoxvirus genes and proteins according to lymphadenopathy24. Rashes appear following the onset
publicly available reference genomes: NC_063383 for of fever, beginning on the face, tongue and oral cavity
the West African MPXV strain, NC_003310.1 for the (enanthem) before spreading across the body. In the later
Central African MPXV Zaire strain and NC_006998 stages of infection, lesions in the oral cavity can cause
for the vaccinia virus (VACV) Western Reserve strain. difficulties in drinking and eating4. However, in the
MPXV genes/proteins are referenced to the Central recent outbreaks, several atypical clinical observations
African MPXV Zaire strain, unless otherwise stated. have been reported. In patients who are MSM, these
include the presence of genital lesions that subsequently
MPXV genome and phylogeny spread to other sites in the body, as well as anal ulcers,
Computational analysis of the Central African MPXV and it appears that skin lesions may be more limited in
Zaire strain revealed the presence of at least 190 open distribution than reported in previous outbreaks24–26.
reading frames (ORFs)15–17. Genes known to be impor- Disease severity can be classified using the lesion
tant for orthopoxviruses are present within the central count, as higher lesion counts correlate with increased
region of the MPXV genome (between ORFs C10L and risk of severe complications4. Patients with severe com-
A25R)16,17. However, a subset of ORFs are either missing plications may experience respiratory and gastrointes-
or truncated in the MPXV genome compared with the tinal issues27, encephalitis4, septicaemia27,28 and ocular
genomes of other orthopoxviruses11,16,18. Disruptions to infections leading to permanent vision loss29. Skin
several ORFs that code for genes involved in immune lesions also increase the likelihood of dermal bacterial
evasion have been reported in the West African clade, infections, especially in patients who are not vaccinated
and these may be responsible for this clade’s lower against smallpox4.
virulence relative to the Central African clade6,11,18. Lesions typically progress through four stages —
The virus exists in two distinct infectious forms, the macular, papular, vesicular and pustular — before
intracellular mature virus and the extracellular enveloped falling off as scabs 1. Patients are typically consid-
virus, which differ in their surface glycoproteins and ered non-contagious once lesions have crusted over.
infect cells via different mechanisms19. MPXV replication However, scabs have been reported to contain significant
is a complex process but is generally thought to be identi- quantities of MPXV DNA even after falling off30, which
cal to other orthopoxviruses19. Entry receptors for MPXV may indicate the presence of infectious viral material.
have not been clearly identified, although it was suggested Of note, viable VARV has been isolated from scabs of
that viral entry is dependent on the viral strain and host patients with smallpox31.
cell type and involves multiple surface receptors, includ- During pregnancy, MPXV can be vertically transmit-
ing chondroitin sulfate20 or heparan sulfate21,22. In VACV, ted from the mother to the fetus32. In one study involving
surface proteins H3, A27 and D8 have been associated four pregnant women who were infected with MPXV in
with viral binding20–22. Following binding, VACV gains the DRC, only one gave birth to a healthy infant. Two
entry into the cell through 11 conserved proteins, which women experienced miscarriage in the first trimester
form a complex known as the entry fusion complex23. and one had a stillbirth. In the stillborn, skin lesions
were observed across the body32. In another study, four
Monkeypox pathology out of five women who were infected with MPXV in the
MPXV infection has an incubation period of 5–21 days, DRC had fetal demise and lesions were observed on
and common symptoms include fever (between the maternal surface of the placenta30. The studies did
38.5 °C and 40.5 °C), headache and myalgia. A distin- not report which clade of MPXV these patients were
guishing feature of MPXV infection is the presence of infected with, although given the location of the studies
swelling at the maxillary, cervical or inguinal lymph it is very likely the Central African clade30.
nodes (lymphadenopathy)1,4. In the recent outbreak, a A study conducted in Zaire during 1980–1985
reported a higher incidence of fatal disease in young chil-
Author addresses dren infected with MPXV, with a case fatality rate of 14.9%
1
A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research in children aged between 0 and 4 years compared with
(A*STAR), Singapore, Singapore. a rate of 0% in individuals aged 10 years or older4. This
2
National Public Health Laboratory, Singapore, Singapore. disparity could potentially be due to differences in their
3
National Centre for Infectious Diseases, Singapore, Singapore. immune responses4. Currently, data on severity of infec-
4
Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, tion in children infected with the West African clade are
National University of Singapore, Singapore, Singapore. lacking33. Nevertheless, the severe outcomes of monkey-
5
Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, pox in pregnant women and in young children highlight
Singapore. the importance of future public health efforts to limit the
6
Tan Tock Seng Hospital, Singapore, Singapore. spread of MPXV and minimize the risk of adverse events.
7
Yong Loo Lin School of Medicine, National University of Singapore, Singapore,
Singapore.
8
School of Biological Sciences, Nanyang Technological University, Singapore, Singapore. MPXV pathogenesis
9
National Institute of Health Research, Health Protection Research Unit in Emerging and The clinical outcome of orthopoxviral infection in a
Zoonotic Infections, University of Liverpool, Liverpool, UK. vertebrate host is strongly dependent on the entry route
10
Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, used by the virus to establish the primary infection34
Liverpool, UK. (Fig. 2). For several orthopoxviruses, such as the highly
0123456789();:
Reviews
0123456789();:
Reviews
Number
of cases
50
100
250
500
1,000
2,000
3,000
4,000
5,000
Fig. 1 | Geographical distribution of confirmed and suspected monkeypox cases during the outbreak between
May and August 2022. Data presented as of 5 August 2022 were obtained from Global.health. Diagram generated with
Datawrapper.
skin infection suggested that dendritic cell migration form of close contact with the infectious pustules that are
from the skin to draining lymph nodes is impaired on symptomatic of monkeypox.
VACV56. The relocation of the virus from the skin to the
lymphatics might also be supported by other mecha- Immunity to MPXV
nisms such as direct lymphatic vessel access, as observed Even though the virus was identified decades ago, human
in skin infection models of Zika virus57. immunity to MPXV infection has not been extensively
Sexual transmission of monkeypox has been characterized. As such, inferences on MPXV interaction
speculated52, and MPXV was recently identified in the with the host immune system are often drawn from
semen of three male patients in Italy26. Cases of mon- studies performed with VACV and related orthopoxvi-
keypox with exclusive genital lesions have also been ruses. In the following sections, we highlight the poten-
reported58, which might indicate preferential MPXV tial mechanisms of host immunity against MPXV and
tropism into the testes. Being an immune-privileged discuss the immune evasion strategies utilized by MPXV
tissue59, the testes could act as a site for latent MPXV during active infection.
infection, but this requires further investigation.
Nonetheless, recent animal studies showed that the Innate immune responses to MPXV. Innate immune cells
related VACV exhibits tropism for testicular and ovar- typically act as the first line of defence following active
ian tissues60,61. Viral shedding was also reported in faeces viral infection, but these cells also serve as targets for
and contact with the rectal mucosa might increase the some viruses. Numerous in vitro and in vivo studies have
likelihood of MPXV transmission35. It was previously demonstrated that monocytes are the initial targets of
noted in patients with human immunodeficiency virus poxviruses36,65–68. Early detection of poxvirus antigen in
1 (HIV-1) that these tissues could act as a reservoir for both monocytes and neutrophils has been suggested to
the virus62. A recent study63 determined that the rectal be a strong predictor of MPXV lethality69. Susceptible
mucosa immune environment in MSM was significantly monocytes are actively recruited to sites of infection, with
different compared with individuals who are heterosex- marked expansion of CD14+ monocytes in the lungs of
ual, with a higher presence of immune activities indic- cynomolgus macaques experiencing viral pneumonia fol-
ative of mucosal injuries. This condition could lead to lowing MPXV infection70. Mouse CD45+CD11b+GR-1int
the recruitment of immune cells, which could then be inflammatory monocytes have also been shown to be
easily targeted by infectious agents, such as observed permissive to VACV replication and may be plausible
with HIV-1 (ref.64). This could similarly apply to MPXV vehicles for virus dissemination47. It was also reported
transmission and infection in MSM. However, this does that human primary M2-like macrophages allowed
not indicate that monkeypox has become a sexually VACV replication and dissemination71. Following VACV
transmitted disease, as MPXV can spread through any infection, these primary macrophages formed actin tails,
0123456789();:
Reviews
Genetic accordions
cell linkages, lamellipodia and branching structures asso- chemokine receptors such as CXCR3, CCR5, CCR6
Multistep events involving gene ciated with the VACV virions, indicating that these cells and CCR7 on these cells was also reported37. Moreover,
application and mutations that might participate in virus spread71. However, it was also natural killer cells isolated from both lymph nodes and
allow poxviruses to subvert reported that depletion of phagocytic cells did not abol- blood were reported to lose their ability to degranulate
host antiviral responses.
ish spread of VACV72 in infected mice, suggesting that and to secrete IFNγ and TNF37. Although no correlation
monocytes and macrophages are not the only immune between viral clearance and natural killer cell numbers
cells that are capable of facilitating virus dissemination. and activities was reported in this NHP model37, the
Nevertheless, Ly6G+ innate immune cells (both neutro- importance of natural killer cells in controlling MPXV
phils and Ly6G+ monocytes72) were responsible for infil- viral load was demonstrated in CAST/EiJ mice. This
trating and controlling virus-infected cells, thus limiting strain is exceptionally susceptible to orthopoxvirus
viral tissue damage47,72. These results were indirectly infection76 owing to low numbers of natural killer cells.
confirmed by a study that found an association between IL-15 treatment protected CAST/EiJ mice from lethal
low numbers of blood neutrophils with moribundity in MPXV infection even when both CD4+ and CD8+ T cells
MPXV-infected animals73. It is also worth noting that were depleted76. This implies that the expanded natural
immune cells recruited to the site of infection control killer cells were responsible for the protective effect as
only local pathogenesis and tissue pathology, but not treatment with IL-15 is known to transiently increase
virus dissemination, and a systemic immune response the numbers of IFNγ-secreting natural killer cells and
is required to prevent widespread infection74. CD8+ T cells77.
Natural killer cells are an important component of Similarly, natural killer cells are also needed to con-
innate immunity and, similar to monocytes, are capa- trol both Ectromelia virus (ECTV) and VACV infection
ble of shaping the adaptive immune response75. In in C57BL/6 mice78,79. ECTV is a natural orthopoxviral
MPXV-infected rhesus macaques, numbers of natural pathogen of mice, and is often used to induce experi-
killer cells expand significantly in both peripheral blood mental mousepox as a model for other clinically impor-
(a mean 23-fold increase by day 7 post infection) and tant orthopoxviruses80. Interestingly, it was suggested
lymph nodes (a mean 46.1-fold increase by days 8–9 that the expression of CD94 on natural killer cells in
post infection)37. Prior to this rapid proliferation, the C57BL/6 mice is absolutely essential for conferring
migratory capacity of the various natural killer cell resistance to ECTV infection81. This is mediated by
subsets was significantly impaired by MPXV infection, the NKG2E and CD94 receptors on natural killer cells,
which severely affected their recruitment into the lym- which bind complexes of MHC class I with the peptide
phoid and/or inflamed tissues37. A downregulation of Qa-1b, which are expressed by infected cells81. NKG2D
has also been reported to participate in natural killer
cell-mediated protection against ECTV infection79 and
Box 2 | Genetic changes in current circulating MPXV it was postulated that CD94-NKG2E and NKG2D may
Understanding whether the recent monkeypox outbreaks are a result of genetic have synergistic activity in inducing optimally protec-
changes in monkeypox virus (MPXV) remains a research challenge. The 2022 tive natural killer cells81. However, the exact mecha-
outbreak is caused by lineage B.1 of the West African clade (MPXV Clade 3)242. At least nisms behind this apparent synergism remain to be
46 single-nucleotide polymorphisms have been found to be specific for this lineage, elucidated, and further studies will be required to better
including 24 non-synonymous single-nucleotide polymorphisms243. Recent molecular understand the roles of natural killer cells in mousepox
epidemiology studies have shown a higher than expected rate of genomic variance infections. Given that CD94 and NKG2 are highly con-
among the outbreak sequences, suggesting accelerated evolution242 and, potentially, served between humans and rodents82, these receptors
APOBEC3 editing244. One preprint publication (not peer reviewed) reports the presence may also play a protective role against MPXV infection
of insertions and deletions in the DNA of the MPXV strain that is circulating in the
in humans.
2022 outbreak compared with MPXV strains that were circulating prior to 2017 and
suggests that these changes may be responsible for the current outbreak14. However,
In humans infected with MPXV, the roles of many innate
it remains unclear whether and how these genetic differences drive the epidemiological immune cells — including monocytes/macrophages,
phenotype. A leading hypothesis proposes that the three non-synonymous single- neutrophils, natural killer cells, conventional dendritic
nucleotide polymorphisms (D209N, P722S and M1741I), which are found in the surface cells, plasmacytoid dendritic cells and innate lymphoid
glycoprotein B21R, a key antibody target, are enhancing the transmissibility of the cells — are currently unknown. The characterization and
virus243,245. However, MPXV is a complex virus, with a DNA-based genome that is profiling of these immune cells during MPXV infection
approximately six times as large (~197 kb) compared with the RNA-based genome of will be essential for understanding their functions and
SARS-CoV-2. It encodes more than 190 open reading frames (ORFs), many of which identifying important biomarkers for disease prognosis.
do not have well-defined functions15–17. Identifying the potential impact of particular VARV infection in animal models triggers systemic
genetic mutations on the viral phenotype is therefore complex and time-consuming.
cytokine responses that correlate with disease outcomes.
That said, the DNA-based genome of MPXV has a much greater capacity to repair
errors incurred during viral replication246 compared with the RNA-based coronaviruses.
In unvaccinated cynomolgus macaques, significant
However, the related orthopoxvirus vaccinia virus (VACV) is capable of ‘phenotypic changes in host gene expression were detected follow-
mutations’ in the form of genetic accordions (multistep events involving gene application ing infection with VARV66. In particular, transcription
and mutation(s) that allow poxviruses to subvert host antiviral responses)247. Specifically, of a cluster of interferon-associated genes was upreg-
a variant of VACV was identified that has a significant amplification of the K3L gene, ulated; this cluster was enriched for genes regulated
which, together with a beneficial point mutation in the same gene, allows the virus by both type I and type II interferons, including PKR,
to largely overcome the host protein kinase R (PKR)-mediated antiviral responses247. STAT1, STAT2, MX1, MX2, IP10, OAS1, OAS2 and OAS3
Such genetic accordions have not been reported in MPXV and further investigations (ref.66). The animals that succumbed to the infection
are needed to investigate whether such a phenomenon could be a plausible mechanism (two out of seven) had minimal interferon responses,
for MPXV evolution and spread.
indicating that this early interferon response protects
0123456789();:
Reviews
MPXV
Ciliated cells
Mucosa
Dendritic Macrophage
cell
Infected
macrophage
h Aerosolized MPXV
Direct virus
Infected access?
dendritic cell
Lymphatic
vessel
Infected
macrophage
Infected
Dendritic cell
c Draining f Infective pustules
lymph node
Natural
killer cells
b Skin route
e Sexual route
Wound MPXV
detected
in semen
Langerhans cell
Epidermis
Infected
fibroblast
d
Hepatocyte
Direct
virus
access?
Macrophage
Dendritic
cell
Lymphatic
Dermis vessel
Kupffer cell
against fatality66. Human IFNβ has been demonstrated VARV and other orthopoxviruses harbour genes that
to inhibit MPXV replication and spread83. However, can modulate TNF84,85 and NF-κB86,87 pathways.
MPXV does not robustly activate TNF-regulated and Although host immunity is required to combat infec-
NF-κB-regulated genes, especially in animals that suc- tions, aberrant immune signalling can adversely affect
cumb to infection66. This is not surprising, given that infection outcomes. In another study of VARV-infected
0123456789();:
Reviews
◀ Fig. 2 | Immunopathogenesis of human monkeypox. a–h | Monkeypox virus (MPXV) B cell and antibody protection. The importance of B cells
might enter the body via the respiratory (panel a) or skin (panel b) route. In the respiratory and immunoglobulins against poxviruses was first
tract, the virus can infect airway epithelial cells such as ciliated cells. Antigen-presenting demonstrated with the successful global vaccination cam-
cells such as dendritic cells and macrophages (MΦ) are also susceptible to MPXV paign that eradicated smallpox, which used a live VACV
infection. Upon inoculation in the skin, the virus infects keratinocytes and fibroblasts.
vaccine94,95. It was further demonstrated that treatment
Skin-resident immune cells such as Langerhans cells, dendritic cells and macrophages
are also targeted. In both scenarios (panels a and b), it is hypothesized that infected
with vaccinia immune globulin (VIG) isolated from the
antigen-presenting cells travel to nearby draining lymph nodes and facilitate its spread serum of vaccinees successfully protected close contacts
through the lymphatic system (panel c). Direct viral access to the lymphatics has been of patients with smallpox from infection96. In rhesus
also speculated. A common feature of human monkeypox is swelling of lymph nodes macaques, VACV-specific B cell responses were instru-
(lymphadenopathy). The abnormal proliferation and retention of natural killer cells might mental in protecting against a lethal MPXV infection53.
be one of the causes. Following its spread through lymphoid tissue, MPXV may target Importantly, epidemiological studies have further
other large organs such as the spleen and liver (panel d). Of note, MPXV antigens have demonstrated that the VACV vaccine confers protection
been previously been detected in both hepatocytes and Kupffer cells in non-human against other poxviruses, including MPXV97. The VACV-
primate (NHP) models. The viraemia wave could then allow the virus to further spread to specific memory B cells and antibody levels induced by
distant organs such as the skin and gonads. Recently, MPXV was isolated from semen of
vaccination were exceptional, in some cases lasting longer
infected individuals, highlighting the possibility of sexual transmission (panel e). The
infection of skin and mucosae leads to the appearance of infective pustules (panel f) and than 50 years98,99. However, only ~50% of vaccinated indi-
ulcers (panel g). The latter release high quantities of virus into the saliva, which viduals at >20 years after vaccination65 had neutralizing
potentially leads to aerosolized transmission of MPXV (panel h). antibody titres greater than 1:32, a correlate of protec-
tion suggested to confer protective immunity against
smallpox100. It is likely that cross-protective immunity
cynomolgus macaques, the levels of IL-8, CCL2 (also against monkeypox may similarly wane over time.
known as MCP1), CCL4 (also known as MIP1β), Fourteen MPXV proteins have been shown to be
IL-6 and IFNγ were significantly increased during recognized by cross-reactive VACV-induced immuno
the first 4 days post infection67. These cytokines drive globulins from human vacinees101. Three proteins in
monocytosis88, which might facilitate enhanced virus particular — MPXV (Zaire-1979_005) proteins D8, H3 and
dissemination through monocytic cell-a ssociated A26 — were targeted by neutralizing antibodies against
viraemia67. Importantly, the macaques eventually suc- MPXV in infected macaques101. In VACV, orthologue D8
cumbed to VARV infection, where high levels of these and H3 proteins are involved in the attachment of mature
cytokines might have contributed to a ‘cytokine storm’ virions22, whereas A26 associates with A27 (ref.102) to
leading to their demise67. Likewise, the levels of IL-1RA, bind surface laminin103. MPXV (Zaire-1979_005) pro-
IL-2, IL-6, IL-8, IFNγ, CCL2, CCL5 (also known as teins C19, A33 and A44 were also prominent antigens
RANTES), G-CSF, GM-CSF and sCD40L were found to for IgM isolated from MPXV-infected macaques dur-
be elevated in MPXV-infected cynomolgus macaques70. ing the acute phase of infection — these proteins could
Furthermore, a relative expansion (0.97-f old to thus be further developed as antigen-based serological
16.3-fold) of CD14+ monocytes was reported during diagnostic tools101. In another study, prophylactic treat-
acute infection, suggesting that the general immune ment with a cocktail of two mAbs — c7D11 and c8A —
milieu promoted the development and recruitment of successfully protected marmosets against lethal MPXV
monocytes following MPXV infection70. infection104. C7D11 and c8A target the VACV proteins L1
In reported cases of human MPXV infection, numer- (ref. 105) and B5 (ref. 106) , respectively, and have been
ous cytokines are elevated following infection (regard- recently formulated as a potential mRNA vaccine encap-
less of disease severity) — these include IL-1β, IL-1RA, sulated in lipid nanoparticles107. Gilchuk et al.108 showed
IL-2R, IL-4, IL-5, IL-6, IL-8, IL-13, IL-15, IL-17, CCL2 that a mixture of human-derived mAbs targeting the
and CCL589. However, in patients with serious disease VACV proteins D8, H3, A33, A27, B5 and L1 effectively
(defined as having >250 lesions), concentrations of cross-neutralized four clinically relevant orthopoxvi-
IL-2R, IL-10, GM-CSF and CCL5 were higher compared ruses, including MPXV and live VARV. However, despite
with those in patients with less severe disease, whereas knowledge of the MPXV proteins that are recognized by
the concentration of pro-inflammatory IL-6 was lower89. neutralizing antibodies, MPXV-specific epitopes (both
This cytokine profile suggests a dominant T helper conformational and linear) have not been extensively
2 cell response characterized by higher levels of IL-4, characterized.
IL-13 and IL-10. Likewise, the reduced levels of IL-2, The isotype composition of the anti-MPXV response
TNF, IFNα and IFNγ also suggest an anti-inflammatory may provide an important clue to pre-existing immunity
microenvironment involving regulatory T cells89. and protection, as IgM antibodies typically dominate in
VACV can evade immune responses by downregulat- primary immune responses, whereas IgG antibodies
Monocytosis ing inflammatory and antiviral immune responses44,90,91, dominate in secondary immune responses. In a cohort
An elevated number of
and MPXV may use a similar strategy to subvert host of 200 patients infected with MPXV who were recruited
infection-fighting monocytes in
the blood circulation. immunity 92. Given that immune mediators often between March 2007 and August 2011 in the DRC, indi-
facilitate crosstalk between immune cells 93, future viduals with both IgM and IgG responses were 5.09 times
Cytokine storm studies should identify the functional relationships more likely to develop severe lesions compared with
A sudden increase in levels of between cytokine profiles and immune cells in order individuals who had IgG-only responses30. Similarly, in a
circulating cytokines leading to
an over-activated immune
to clarify the mechanisms of pathogenesis and deter- cohort of infected individuals from the 2003 MPXV out-
response, which can be mine immune correlates of protection during MPXV break in the United States, patients with moderate/severe
life-threatening. infection. disease had an overall higher titre of anti-orthopoxvirus
0123456789();:
Reviews
IgM 109 compared with those with mild disease, activation of CD8+ T cells114. In a mouse model of VACV
and anti-orthopoxvirus IgG responses were much respiratory infection, IFNγ secretion by primary acti-
reduced and less frequent in patients with moderate/ vated effector CD8+ T cells was shown to protect against
severe disease109. It is plausible that an IgG-only response lethality115. Indeed, CD8+ T cell-derived IFNγ was suffi-
reflects robust levels of cross-protective IgG+ memory cient for protection even in the absence of CD4+ T cells
B cells that produce a dominant secondary antibody and B cells115, highlighting the possibility that CD8+
response, whereas the lack of such memory necessitates T cells also confer protection against infection with other
an IgM-dominated primary response that is less effec- orthopoxviruses. Likewise, memory CD8+ T cells, which
tive at preventing disease. Thus, IgM responses may be a are induced following VACV immunization, were also
biomarker for disease severity. This also emphasizes the demonstrated to protect against lethal ECTV infection
critical and immediate need to extensively characterize in mice116. These memory CD8+ T cells execute their
the antibody profile of patients with MPXV in different protective effects via a combination of IFNγ and per-
cohorts. Likewise, VACV vaccine correlates of protection forin secretion, and work concomitantly with primary
must be determined to explain why certain vaccinated effector CD8+ T cells to achieve optimal protection116.
patients experience breakthrough infections. In humans, standard smallpox vaccine administered by
scarification117 was also able to induce primary cyto-
T cell immunity. CD4+ T cells, particularly T follicular toxic CD8+ T cells and IFNγ-producing T cells118. This
helper cells, play a role in enhancing recall and differ- observation was supported by another study, in which
entiation of memory B cells into antibody-secreting participants received live vaccinia smallpox vaccine
cells110. Memory CD4+ T cells were found to persist for (Dryvax)119. High levels of IFNγ-producing CD8+ and
up to 50 years or longer following VACV vaccine, with an CD4+ T cells were detected following immunization119.
estimated half-life of 8–15 years99. These VACV-specific In humans infected with VACV, activated CD4+ T cells
CD4+ T cells were capable of producing IFNγ and TNF were shown to upregulate genes related to cytolytic
following stimulation99. However, no direct correlation activities120. Interestingly, MHC class II-restricted cytol-
was reported between numbers of virus-specific CD4+ ytic CD4+ T cells have also been reported in individuals
T cells and anti-VACV antibody titres99. By contrast, immunized with VACV121. These cells could be respon-
the number of CD4+ T cells was shown to be critically sible for virus clearance in vaccinees with reduced or
important in inducing a protective antibody response missing memory CD8+ T cell responses99. In the experi-
against lethal MPXV infection in VACV-vaccinated mental mousepox model, perforin-dependent cytolytic
rhesus macaques53. Simian immunodeficiency virus CD4+ T cells have been reported122. Taken together, these
(SIV)-infected macaques with CD4 + T cell counts observations highlight the importance of T lymphocytes
<300 cells mm–3 were not able to produce VACV-specific in controlling orthopoxviral infections.
IgG following vaccination and died when challenged with Across the orthopoxvirus proteome, numerous
MPXV53. This observation is of high concern to both CD4+ and CD8+ T cell epitopes have been identified
VACV-vaccinated and unvaccinated individuals with in humans, mice and NHPs123–125. Many are conserved
uncontrolled HIV-1 infection, as their CD4+ T cell counts among the major orthopoxviruses and bind to human
are typically very low111. This group of individuals is there- MHC class I and class II molecules126–128. In particu-
fore at high risk of developing severe MPXV infection if lar, CD8+ T cells specific for two identified epitopes
exposed112. They might also experience a more compli- (MHC class II-restricted GRVFDKADGKSKRDA
cated pathology and provide the virus with an opportu- and MHC class I-restricted NPVTVINEY) in the immediate-
nity to acquire mutations that result in higher virulence early E3 protein of VACV were capable of killing infected
or transmission potential113. By contrast, a recent patient cells and halting the spread of VACV129. Both epitopes are
infected with MPXV who was on antiretroviral treat- conserved in the MPXV homologue, which is encoded
ment for HIV-1 had a CD4+ T cell count >700 cells mm–3 by the MPXV F3L gene130. An earlier study showed that
and did not experience a severe disease outcome25. VACV missing the E3 protein did not protect cynomol-
This may suggest an important role for CD4+ T cells gus macaques from subsequent MPXV infection131. As E3
in regulating monkeypox severity. Nevertheless, more protein is detected within 30 min of VACV infection132,
studies will need to be carried out to fully understand it should be readily processed and presented by infected
the role of CD4+ T cells during MPXV infection. cells, allowing T cell-mediated lysis at an early infection
In addition to supporting antibody development, stage before virion production and release. These prop-
T cells can play direct antiviral roles. Given that erties make E3 an excellent possible candidate for future
orthopoxviruses, including MPXV, infect and dissem- vaccine designs targeting all major orthopoxviruses.
inate in macrophages36,65–68, cytolytic T cells can be Despite the potential importance of T cells in dis-
instrumental in killing infected macrophages to prevent ease protection, smallpox vaccination does not neces-
viral spread. CD8+ T cells have been shown to eradicate sarily provide robust T cell-mediated immunity against
virus-infected monocytes and minimize virus dissemi- MPXV. In two out of five vaccinated individuals who
nation in a mouse model of VACV infection47. In fact, subsequently contracted MPXV, orthopoxvirus-specific
activation of CD8+ T cells in response to VACV infection CD4+ and CD8+ T cells were not detectable throughout
has been shown to be dependent on γδT cells, which convalescence 109 . Furthermore, similar levels of
present VACV peptides via MHC class I molecules114. orthopoxvirus-specific CD4+ T cell reactivity were
Moreover, γδT cells also upregulate co-stimulatory mol- observed in vaccinated and unvaccinated patients, and
ecules CD80 and CD86 and secrete IL-1 and IFNα for orthopoxvirus-specific CD8+ T cell responses were in
0123456789();:
Reviews
0123456789();:
Reviews
regulation of the intrinsic apoptotic pathway. Numerous VACV is considered the virus’s most potent inhibitor of
orthopoxvirus-encoded serine protease inhibitors (SPIs; apoptosis154. CrmA interferes with granzyme B155, which
serpins) have also been reported85,133,152,153, such as CrmA is secreted by cytotoxic T cells to initiate cell death in
in cowpox virus (CPXV)153; its homologue SPI-2 (B13) in the virus-infected target cells156. It also inhibits caspase 1
0123456789();:
Reviews
0123456789();:
Reviews
Box 3 | Ring-fencing vaccination against monkeypox little supporting clinical data from humans. The anti-
viral drug tecovirimat does not affect the intracellular
To halt the potential spread of monkeypox virus (MPXV) globally, ring vaccination has form of the virus (the intracellular mature virus), but
been implemented in several countries including the United Kingdom, the United targets the VP37 membrane protein of MPXV to prevent
States and Canada. This usually involves vaccinating people who may have been the formation of enveloped virions that are capable of
exposed to the virus through interactions with an infected person. Vaccination is
cell egress, thus disrupting viral spread188. Tecovirimat
performed using smallpox vaccines, which are thought to offer protection against
MPXV. Some of the challenges facing a successful ring vaccination are discussed below. was effective at reducing monkeypox severity in NHP
• Contact tracing: to identify all individuals who are potentially exposed, extensive models189,190, but its effectiveness is reduced if treatment
contact tracing and robust testing needs to be carried out. For logistic reasons this may is given more than 5 days post challenge191. This sug-
not be possible in every country. Furthermore, given the current characterization of gests a caveat if tecovirimat is given based on the onset
non-endemic monkeypox as a disease that spreads primarily within the community of symptoms, which may appear much later following
of men who have sex with men (MSM), patients may be reluctant to come forward for infection1,4. Nevertheless, a small study used tecovirimat
diagnosis, especially in countries where homosexuality is stigmatized or criminalized248. in a single patient infected with monkeypox, who then
• Efficiencies of second-generation and third-generation vaccines: past data showed showed a shorter duration of viral shedding and illness
that smallpox vaccination was 85% protective against MPXV60. However, this compared with six untreated patients28.
value was obtained from individuals vaccinated with the first-generation vaccine. Another smallpox therapeutic is brincidofovir, which
Nevertheless, currently available second-generation and third-generation vaccines
is approved by the FDA but not the European Medicines
were also reported to be approximately 85% protective against MPXV in animals
models249,250, but there are no data on their efficacy in humans exposed to MPXV.
Agency (EMA). Brincidofovir inhibits orthopoxvi-
rus DNA polymerase-mediated DNA synthesis. It has
• Willingness to comply: not everyone identified will be willing to receive the vaccine.
In a recent report, only ~14% of community contacts and ~69% of potentially exposed shown efficacy against monkeypox in prairie dog and
health-care workers in the United Kingdom were willing to be vaccinated40. Moreover, mouse models192,193. In the same small study discussed
the stigmatization of potentially being associated with the community of MSM might above28, three patients were treated with brincidofovir,
further deter vaccination efforts. but all three ceased therapy owing to toxicity (elevated
liver enzymes). Cidofovir28, the active drug form of brin-
protection, the efficacy of a single dose of JYNNEOS cidofovir, also shows anti-poxvirus activity in vitro and
may be similar to ACAM2000 within the first critical in animal studies194, but it is nephrotoxic195–197.
weeks of post-exposure prophylaxis. In the same prairie VIG showed promising activity in preventing
dog study referenced above, post-exposure vaccination smallpox in exposed individuals198 and has exhibited
with JYNNEOS resulted in 38–88% survival181. cross-neutralizing activity against MPXV in rhesus
A critical aspect of vaccine development is under- macaques53. However, neither VIG nor other mAbs
standing the mechanisms by which different vaccines or antibody cocktails have yet been studied against
elicit immune responses, as different formulations can monkeypox in the clinic.
induce either specific B cell-mediated or T cell-mediated
immunity184. Likewise, the immunocompetency of vac- Concluding remarks
cine recipients as well as the route of immunization Immunotherapeutics and preventive strategies are
will also affect the quality of the immune response184. important public health tools that complement strin-
Given the frequent mucosal route of infection, the spe- gent contact tracing in curbing the spread of monkey-
cific induction of mucosal immunity may be crucial in pox. Similarly, serology-based diagnostics are valuable
the context of MPXV, as documented for other infec- surveillance tools for contact tracing and understanding
tious pathogens that transmit via the mucosal route185. exposure history. However, such serological diagnostic
Currently, it is not well understood whether any of the tools must be specific for MPXV, given the baseline
available vaccines are able to induce mucosal immunity, of vaccine-induced poxvirus immunity199. Expansion of
and this remains a key future research need. surveillance networks and identification of surveillance
Another research need is to understand whether there gaps are also critical for a successful ring-fencing strat-
is a risk of creating new virus strains when individuals egy (see Box 3 for details). Notably, there is a major need
infected with monkeypox are vaccinated. Co-infecting in public health to better inform potentially exposed
poxviruses can evolve by exchanging genetic information persons of the benefits and risks of vaccination.
via homologous or non-homologous recombination186. Historically, the collection of clinical data on mon-
It is currently unknown how frequently genes from live keypox has been hindered by the sparseness and unpre-
or attenuated poxvirus-based vaccines may recombine dictability of cases in endemic countries, impeding
with monkeypox. accurate risk–benefit calculations in managing mon-
In addition to the VACV or MVA-based vaccines, keypox. The current outbreaks provide an opportunity
newer genetically engineered VACV or other poxvirus to evaluate current and novel treatments and vaccine
strains such as NYVAC and ALVAC have also been experi- options and to establish correlates of vaccine protection.
mented with as potential vaccine vectors for other diseases. Notably, the ring vaccination trial design can provide high
Ring vaccination trial design However, they generally induce lower antibody titres187, statistical power with relatively few subjects owing to
A recently developed approach and their effectiveness against monkeypox is unclear. the high potential attack rate200. However, in generaliz-
that recruits subjects linked to ing such findings, caution must be taken to account for
an infected case and allows Therapeutics variance in the route of transmission.
for the simultaneous evaluation
of vaccine efficacy and
Several therapeutics that were developed for smallpox Several key research questions remain to be answered.
effectiveness during ongoing are available for monkeypox, but their effectiveness Studies of the human systemic and mucosal immune
outbreaks. for monkeypox is based on preclinical evidence with responses during MPXV infection are currently limited
0123456789();:
Reviews
and will be required to better understand the mecha- recommended by the UK Health Security Agency that
nisms of immune defence against MPXV. Importantly, vaccination should be given to men who are homosexual
it is not currently known whether prior infection with or bisexual who are at a higher risk of exposure to help
VACV or MPXV, or smallpox vaccination, induces any control the current outbreak205.
form of mucosal immunity. It will be critical to char- Many other infectious diseases are endemic to
acterize the mucosal immune responses given that Africa206, and co-infection with MPXV is possible.
MPXV and other poxviruses can be transmitted via For instance, co-infection of alphaviral infections and
respiratory aerosols1. Knowledge of the roles of IgA malaria can significantly modulate host immunity
and tissue-resident memory T cells in MPXV infection and affect infection outcomes207–209. Given the current
would be particularly critical, enabling a deeper under- transmission among MSM in non-endemic countries,
standing of MPXV-related respiratory complications22. there is also a need to understand monkeypox disease
Likewise, the preputial mucosal immunity should also and vaccination in the context of co-infection with other
be characterized, as MPXV DNA has been identified in diseases that have a disproportionate burden in the com-
semen26. munity of MSM; this is especially true for HIV-1, which
Defining the immunological correlates of protection can severely impair adaptive immune responses210.
is also an important goal for the evaluation of newer Risk factors for severe MPXV should also be identi-
vaccines, especially those targeted towards the vul- fied. Pregnant women, young children and unvaccinated
nerable populations of pregnant women and children. individuals are known to be especially susceptible1,4,30,32.
Therefore, other than being unvaccinated, what other However, other populations also need characteriza-
factors (for example, behavioural, geographical, nutri- tion, including older people, individuals on long-term
tional, medical, immunological or genetic factors) are medications and individuals with underlying metabolic
involved? It was recently reported that the quality of diseases who may manifest the disease differently.
innate immune responses in young children determines Long-term disease sequelae should be tracked in
the severity of respiratory viral infection201. Likewise, patients, including children born to mothers infected
as reported for SARS-CoV-2 infections, children who with MPXV. Experience with the Zika outbreak showed
are infected tend to have reduced B cell202 and T cell203 that children with in utero exposure can develop some
responses compared with adults. The characterization developmental issues later in life211. This research focus
of adaptive immune responses in children infected with should be given more urgency, given that available data
MPXV could clarify why children tend to exhibit a more suggest that only 20–25% of pregnant women infected
severe disease and lower vaccine effectiveness1,4,7. It is with MPXV would successfully give birth30,32. Moreover,
also important to understand the risks of vaccination young children are apparently more susceptible to severe
in vulnerable populations, especially young children monkeypox4. Currently, data on fetal development follow-
and pregnant women1,4,30,32. Clinical studies are lacking ing congenital MPXV infection are lacking. In addition,
in these populations for most of the available thera- longitudinal monitoring of patients with MPXV would
peutics and vaccines. Furthermore, some important also allow the determination of whether infection with
vaccines and therapeutics (for example, ACAM2000 MPXV can lead to long-term effects, as observed after
and brincidofovir) are not recommended or are even infection with SARS-CoV-2 during the current pandemic.
contraindicated for these populations due to increased
potential risk of side effects204. Nevertheless, it was Published online 5 September 2022
1. McCollum, A. M. & Damon, I. K. Human monkeypox. action. Lancet Microbe https://doi.org/10.1016/ 20. Hsiao, J. C., Chung, C. S. & Chang, W. Vaccinia
Clin. Infect. Dis. 58, 260–267 (2014). S2666-5247(22)00153-7 (2022). virus envelope D8L protein binds to cell surface
2. Cho, C. T. & Wenner, H. A. Monkeypox virus. 10. Likos, A. M. et al. A tale of two clades: monkeypox chondroitin sulfate and mediates the adsorption
Bacteriol. Rev. 37, 1–18 (1973). viruses. J. Gen. Virol. 86, 2661–2672 (2005). of intracellular mature virions to cells. J. Virol. 73,
3. Cohen, J. Monkeypox outbreak questions intensify as 11. Chen, N. et al. Virulence differences between 8750–8761 (1999).
cases soar. Science 376, 902–903 (2022). monkeypox virus isolates from West Africa and the 21. Chung, C. S., Hsiao, J. C., Chang, Y. S. & Chang, W.
4. Jezek, Z., Szczeniowski, M., Paluku, K. M. & Congo Basin. Virology 340, 46–63 (2005). A27L protein mediates vaccinia virus interaction with
Mutombo, M. Human monkeypox: clinical features 12. Bunge, E. M. et al. The changing epidemiology cell surface heparan sulfate. J. Virol. 72, 1577–1585
of 282 patients. J. Infect. Dis. 156, 293–298 of human monkeypox — a potential threat? A (1998).
(1987). systematic review. PLoS Negl. Trop. Dis. 16, 22. Lin, C. L., Chung, C. S., Heine, H. G. & Chang, W. Vaccinia
5. Kimball, S. WHO declares rapidly spreading e0010141 (2022). virus envelope H3L protein binds to cell surface heparan
monkeypox outbreak a global health emergency. 13. Minhaj, F. S. et al. Monkeypox outbreak — nine sulfate and is important for intracellular mature virion
CNBC https://www.cnbc.com/2022/07/23/who- states, May 2022. MMWR Morb. Mortal. Wkly. Rep. morphogenesis and virus infection in vitro and in vivo.
declares-spreading-monkeypox-outbreak-a-global- 71, 764–769 (2022). J. Virol. 74, 3353–3365 (2000).
health-emergency.html (2022). 14. Desingu, P. A. & Nagarajan, K. Genomic regions 23. Moss, B. Membrane fusion during poxvirus entry.
6. Reynolds, M. G. & Damon, I. K. Outbreaks of human insertion and deletion in monkeypox virus causing Semin. Cell Dev. Biol. 60, 89–96 (2016).
monkeypox after cessation of smallpox vaccination. multi-country outbreak — 2022. Preprint at 24. Perez Duque, M. et al. Ongoing monkeypox virus
Trends Microbiol. 20, 80–87 (2012). bioRxiv https://doi.org/10.1101/2022.06.28.497936 outbreak, Portugal, 29 April to 23 May 2022.
7. Fine, P. E. M., Jezek, Z., Grab, B. & Dixon, H. (2022). Eurosurveillance 27, 2200424 (2022).
The transmission potential of monkeypox virus in 15. Kugelman, J. R. et al. Genomic variability of monkeypox This study reports the epidemiological and clinical
human populations. Int. J. Epidemiol. 17, 643–650 virus among humans, Democratic Republic of the Congo. characteristics of a group of 27 confirmed cases
(1988). Emerg. Infect. Dis. 20, 232–239 (2014). from Portugal during this current outbreak.
8. Thornhill, J. P. et al. Monkeypox virus infection in 16. Shchelkunov, S. N. et al. Human monkeypox and 25. Hammerschlag, Y. et al. Monkeypox infection
humans across 16 countries — April–June 2022. smallpox viruses: genomic comparison. FEBS Lett. presenting as genital rash, Australia, May 2022.
N. Engl. J. Med. https://doi.org/10.1056/ 509, 66–70 (2001). Eurosurveillance 27, 2200411 (2022).
NEJMoa2207323 (2022). 17. Shchelkunov, S. N. et al. Analysis of the monkeypox This study describes the case report of an
This paper reports on the clinical findings of 528 virus genome. Virology 297, 172–194 (2002). individual returning to Australia from Europe,
patients infected with monkeypox from the current 18. Weaver, J. R. & Isaacs, S. N. Monkeypox virus and who later went on to develop genital lesions arising
outbreak. insights into its immunomodulatory proteins. from MPXV infection.
9. Otu, A., Ebenso, B., Walley, J., Barcelo, J. M. & Immunol. Rev. 225, 96–113 (2008). 26. Antinori, A. et al. Epidemiological, clinical
Ochu, C. L. Global human monkeypox outbreak: 19. McFadden, G. Poxvirus tropism. Nat. Rev. Microbiol. and virological characteristics of four cases of
atypical presentation demanding urgent public health 3, 201–213 (2005). monkeypox support transmission through sexual
0123456789();:
Reviews
contact, Italy, May 2022. Eurosurveillance 27, immune cells in the skin. Cell Host Microbe 13, 70. Johnson, R. F. et al. Comparative analysis of monkeypox
2200421 (2022). 155–168 (2013). virus infection of cynomolgus macaques by the
This paper describes the clinical findings obtained 48. Chahroudi, A. et al. Vaccinia virus tropism for primary intravenous or intrabronchial inoculation route.
from four infected males, all of whom participate in hematolymphoid cells is determined by restricted J. Virol. 85, 2112–2125 (2011).
activities with MSM. expression of a unique virus receptor. J. Virol. 79, 71. Byrd, D. et al. Primary human macrophages serve
27. Huhn, G. D. et al. Clinical characteristics of human 10397–10407 (2005). as vehicles for vaccinia virus replication and
monkeypox, and risk factors for severe disease. 49. Chapman, J. L., Nichols, D. K., Martinez, M. J. dissemination. J. Virol. 88, 6819–6831 (2014).
Clin. Infect. Dis. 41, 1742–1751 (2005). & Raymond, J. W. Animal models of orthopoxvirus 72. Fischer, M. A. et al. CD11b+, Ly6G+ cells produce type
28. Adler, H. et al. Clinical features and management infection. Vet. Pathol. 47, 852–870 (2010). I interferon and exhibit tissue protective properties
of human monkeypox: a retrospective observational 50. Moulton, E. A., Atkinson, J. P. & Buller, R. M. L. following peripheral virus infection. PLoS Pathog. 7,
study in the UK. Lancet Infect. Dis. https://doi.org/ Surviving mousepox infection requires the e1002374 (2011).
10.1016/S1473-3099(22)00228-6 (2022). complement system. PLoS Pathog. 4, e1000249 73. Nagata, N. et al. Pathogenesis of fulminant monkeypox
This paper describes the retrospective clinical (2008). with bacterial sepsis after experimental infection with
and virological findings of seven patients infected 51. Institute of Medicine (US) Committee on the West African monkeypox virus in a cynomolgus monkey.
with MPXV in the United Kingdom between 2018 Assessment of Future Scientific Needs for Live Variola Int. J. Clin. Exp. Pathol. 7, 4359–4370 (2014).
and 2021. Virus. Assessment of Future Scientific Needs for Live 74. Davies, M. L. et al. A systemic macrophage response
29. Learned, L. A. et al. Extended interhuman transmission Variola Virus (National Academies Press (US), 1999). is required to contain a peripheral poxvirus infection.
of monkeypox in a hospital community in the 52. Alakunle, E., Moens, U., Nchinda, G. & Okeke, M. I. PLoS Pathog. 13, e1006435 (2017).
Republic of the Congo, 2003. Am. J. Trop. Med. Hyg. Monkeypox virus in Nigeria: infection biology, 75. Paust, S., Senman, B. & von Andrian, U. H. Adaptive
73, 428–434 (2005). epidemiology, and evolution. Viruses 12, 1257 immune responses mediated by natural killer cells.
30. Pittman, P. R. et al. Clinical characterization of human (2020). Immunol. Rev. 235, 286–296 (2010).
monkeypox infections in the Democratic Republic 53. Edghill-Smith, Y. et al. Smallpox vaccine-induced 76. Earl, P. L., Americo, J. L. & Moss, B. Natural killer cells
of the Congo. Preprint at medRxiv https://doi.org/ antibodies are necessary and sufficient for protection expanded in vivo or ex vivo with IL-15 overcomes the
10.1101/2022.05.26.22273379 (2022). against monkeypox virus. Nat. Med. 11, 740–747 inherent susceptibility of CAST mice to lethal infection
31. Downie, A. W. & Dumbell, K. R. Survival of variola (2005). with orthopoxviruses. PLoS Pathog. 16, e1008505
virus in dried exudate and crusts from smallpox 54. Deng, L. et al. Vaccinia virus subverts a mitochondrial (2020).
patients. Lancet 1, 550–553 (1947). antiviral signaling protein-dependent innate immune 77. Jayaraman, A. et al. IL-15 complexes induce NK- and
32. Mbala, P. K. et al. Maternal and fetal outcomes response in keratinocytes through its double-stranded T-cell responses independent of type I IFN signaling
among pregnant women with human monkeypox RNA binding protein, E3. J. Virol. 82, 10735–10746 during rhinovirus infection. Mucosal Immunol. 7,
infection in the Democratic Republic of Congo. (2008). 1151–1164 (2014).
J. Infect. Dis. 216, 824–828 (2017). 55. Cann, J. A., Jahrling, P. B., Hensley, L. E. & 78. Bukowski, J. F., Woda, B. A., Habu, S., Okumura, K.
This study describes the effect of MPXV infection Wahl-Jensen, V. Comparative pathology of smallpox & Welsh, R. M. Natural killer cell depletion enhances
in pregnant women and reports the maternal and and monkeypox in man and macaques. J. Comp. virus synthesis and virus-induced hepatitis in vivo.
fetal outcomes following infection. Pathol. 148, 6–21 (2013). J. Immunol. 131, 1531–1538 (1983).
33. Centers for Disease Control and Prevention. 56. Aggio, J. B., Krmeská, V., Ferguson, B. J., Wowk, P. F. 79. Fang, M., Lanier, L. L. & Sigal, L. J. A role for NKG2D
Clinical considerations for Monkeypox in children & Rothfuchs, A. G. Vaccinia virus infection inhibits skin in NK cell-mediated resistance to poxvirus disease.
and adolescents. CDC https://www.cdc.gov/poxvirus/ dendritic cell migration to the draining lymph node. PLoS Pathog. 4, e30 (2008).
monkeypox/clinicians/pediatric.html (2022). J. Immunol. 206, 776–784 (2021). 80. Fenner, F. Adventures with poxviruses of vertebrates.
34. Stanford, M. M., McFadden, G., Karupiah, G. & 57. Reynoso, G. V. et al. Lymph node conduits transport FEMS Microbiol. Rev. 24, 123–133 (2000).
Chaudhri, G. Immunopathogenesis of poxvirus virions for rapid T cell activation. Nat. Immunol. 20, 81. Fang, M. et al. CD94 is essential for NK cell-mediated
infections: forecasting the impending storm. 602–612 (2019). resistance to a lethal viral disease. Immunity 34,
Immunol. Cell Biol. 85, 93–102 (2007). 58. Davido, B., D’anglejan, E., Jourdan, J., Robinault, A. 579–589 (2011).
35. Rimoin, A. W. et al. Major increase in human & Davido, G. Monkeypox 2022 outbreak: cases with 82. Vance, R. E., Jamieson, A. M. & Raulet, D. H.
monkeypox incidence 30 years after smallpox exclusive genital lesions. J. Travel. Med. https://doi.org/ Recognition of the class Ib molecule Qa-1b by putative
vaccination campaigns cease in the Democratic 10.1093/jtm/taac077 (2022). activating receptors CD94/NKG2C and CD94/NKG2E
Republic of Congo. Proc. Natl Acad. Sci. USA 107, This paper describes the presence of genital on mouse natural killer cells. J. Exp. Med. 190,
16262–16267 (2010). lesions in recent cases involving young men 1801–1812 (1999).
36. Zaucha, G. M., Jahrling, P. B., Geisbert, T. W., engaging in activities with MSM. 83. Johnston, S. C. et al. In vitro inhibition of monkeypox
Swearengen, J. R. & Hensley, L. The pathology of 59. Forrester, J. V., Mölzer, C. & Kuffova, L. Immune privilege virus production and spread by Interferon-β. Virol. J.
experimental aerosolized monkeypox virus infection furnishes a niche for latent infection. Front. Ophthalmol. 9, 5 (2012).
in cynomolgus monkeys (Macaca fascicularis). https://doi.org/10.3389/fopht.2022.869046 (2022). 84. Shchelkunov, S. N. Orthopoxvirus genes that mediate
Lab. Invest. 81, 1581–1600 (2001). 60. Prow, N. A. et al. A vaccinia-based single vector disease virulence and host tropism. Adv. Virol. 2012,
37. Song, H. et al. Monkeypox virus infection of rhesus construct multi-pathogen vaccine protects against 524743 (2012).
macaques induces massive expansion of natural both Zika and chikungunya viruses. Nat. Commun. 9, 85. Suraweera, C. D., Hinds, M. G. & Kvansakul, M.
killer cells but suppresses natural killer cell functions. 1230 (2018). Poxviral strategies to overcome host cell apoptosis.
PLoS ONE 8, e77804 (2013). 61. Zhao, Y., Adams, Y. F. & Croft, M. Preferential replication Pathogens 10, 6 (2020).
38. Dai, P. et al. Modified vaccinia virus ankara triggers of vaccinia virus in the ovaries is independent of immune 86. Mohamed, M. R. & McFadden, G. NF-κB inhibitors:
type I IFN production in murine conventional dendritic regulation through IL-10 and TGF-β. Viral Immunol. 24, strategies from poxviruses. Cell Cycle 8, 3125–3132
cells via a cGAS/STING-mediated cytosolic DNA-sensing 387–396 (2011). (2009).
pathway. PLoS Pathog. 10, e1003989 (2014). 62. Khoury, G. et al. Human immunodeficiency virus 87. Shisler, J. L. & Jin, X. L. The vaccinia virus K1L gene
39. Engelmayer, J. et al. Vaccinia virus inhibits the persistence and T-cell activation in blood, rectal, product inhibits host NF-κB activation by preventing
maturation of human dendritic cells: a novel mechanism and lymph node tissue in human immunodeficiency virus- IκBα degradation. J. Virol. 78, 3553–3560 (2004).
of immune evasion. J. Immunol. 163, 6762–6768 infected individuals receiving suppressive antiretroviral 88. Dutta, P. & Nahrendorf, M. Regulation and
(1999). therapy. J. Infect. Dis. 215, 911–919 (2017). consequences of monocytosis. Immunol. Rev. 262,
40. Li, P. et al. Disruption of MHC class II-restricted 63. Kelley, C. F. et al. The rectal mucosa and condomless 167–178 (2014).
antigen presentation by vaccinia virus. J. Immunol. receptive anal intercourse in HIV-negative MSM: 89. Johnston, S. C. et al. Cytokine modulation correlates
175, 6481–6488 (2005). implications for HIV transmission and prevention. with severity of monkeypox disease in humans. J. Clin.
41. Humlová, Z., Vokurka, M., Esteban, M. & Mucosal Immunol. 10, 996–1007 (2017). Virol. 63, 42–45 (2015).
Mělková, Z. Vaccinia virus induces apoptosis of infected 64. Mikulak, J., Di Vito, C., Zaghi, E. & Mavilio, D. Host 90. Howell, M. D. et al. Cytokine milieu of atopic
macrophages. J. Gen. Virol. 83, 2821–2832 (2002). immune responses in HIV-1 infection: the emerging dermatitis skin subverts the innate immune response
42. Beauchamp, N. M., Busick, R. Y. & pathogenic role of siglecs and their clinical correlates. to vaccinia virus. Immunity 24, 341–348 (2006).
Alexander-Miller, M. A. Functional divergence Front. Immunol. 8, 314 (2017). 91. van Den Broek, M. et al. IL-4 and IL-10 antagonize
among CD103+ dendritic cell subpopulations 65. Hammarlund, E. et al. Monkeypox virus evades IL-12-mediated protection against acute vaccinia virus
following pulmonary poxvirus infection. J. Virol. 84, antiviral CD4+ and CD8+ T cell responses by infection with a limited role of IFN-γ and nitric oxide
10191–10199 (2010). suppressing cognate T cell activation. Proc. Natl Acad. synthetase 2. J. Immunol. 164, 371–378 (2000).
43. Humrich, J. Y. et al. Vaccinia virus impairs directional Sci. USA 105, 14567–14572 (2008). 92. Thakur, A., Mikkelsen, H. & Jungersen, G. Intracellular
migration and chemokine receptor switch of human 66. Rubins, K. H. et al. The host response to smallpox: pathogens: host immunity and microbial persistence
dendritic cells. Eur. J. Immunol. 37, 954–965 (2007). analysis of the gene expression program in peripheral strategies. J. Immunol. Res. 2019, 1356540 (2019).
44. Liu, L. et al. Vaccinia virus induces strong blood cells in a nonhuman primate model. Proc. Natl 93. Srivastava, R. M., Marincola, F. M. & Shanker, A.
immunoregulatory cytokine production in healthy Acad. Sci. USA 101, 15190–15195 (2004). Editorial: lymphocyte functional crosstalk and
human epidermal keratinocytes: a novel strategy for 67. Jahrling, P. B. et al. Exploring the potential of variola regulation. Front. Immunol. 10, 2916 (2019).
immune evasion. J. Virol. 79, 7363–7370 (2005). virus infection of cynomolgus macaques as a model 94. Strassburg, M. A. The global eradication of smallpox.
45. Walzer, T., Galibert, L. & De Smedt, T. Poxvirus for human smallpox. Proc. Natl Acad. Sci. USA 101, Am. J. Infect. Control. 10, 53–59 (1982).
semaphorin A39R inhibits phagocytosis by 15196–15200 (2004). 95. Cherry, J. D. et al. Clinical and serologic study of four
dendritic cells and neutrophils. Eur. J. Immunol. 68. Rubins, K. H., Hensley, L. E., Relman, D. A. & smallpox vaccines comparing variations of dose and
35, 391–398 (2005). Brown, P. O. Stunned silence: gene expression route of administration. Primary percutaneous
46. Norbury, C. C., Malide, D., Gibbs, J. S., Bennink, J. R. programs in human cells infected with monkeypox vaccination. J. Infect. Dis. 135, 145–154 (1977).
& Yewdell, J. W. Visualizing priming of virus-specific or vaccinia virus. PLoS ONE 6, e15615 (2011). 96. Kempe, C. H. et al. The use of vaccinia hyperimmune
CD8+ T cells by infected dendritic cells in vivo. 69. Song, H. et al. Poxvirus antigen staining of immune γ-globulin in the prophylaxis of smallpox. Bull. World
Nat. Immunol. 3, 265–271 (2002). cells as a biomarker to predict disease outcome in Health Organ. 25, 41–48 (1961).
47. Hickman, H. D. et al. Anatomically restricted monkeypox and cowpox virus infection in non-human 97. Jacobs, B. L. et al. Vaccinia virus vaccines: past,
synergistic antiviral activities of innate and adaptive primates. PLoS ONE 8, e60533 (2013). present and future. Antivir. Res. 84, 1–13 (2009).
0123456789();:
Reviews
98. Crotty, S. et al. Cutting edge: long-term B cell memory 121. Littaua, R. A., Takeda, A., Cruz, J. & Ennis, F. A. 145. Jefferies, C. A. Regulating IRFs in IFN driven disease.
in humans after smallpox vaccination. J. Immunol. Vaccinia virus-specific human CD4+ cytotoxic Front. Immunol. 10, 325 (2019).
171, 4969–4973 (2003). T-lymphocyte clones. J. Virol. 66, 2274–2280 (1992). 146. Colamonici, O. R., Domanski, P., Sweitzer, S. M.,
99. Hammarlund, E. et al. Duration of antiviral immunity 122. Fang, M. et al. Perforin-dependent CD4+ T-cell Larner, A. & Buller, R. M. Vaccinia virus B18R gene
after smallpox vaccination. Nat. Med. 9, 1131–1137 cytotoxicity contributes to control a murine poxvirus encodes a type I interferon-binding protein that blocks
(2003). infection. Proc. Natl Acad. Sci. USA 109, 9983–9988 interferon α transmembrane signaling. J. Biol. Chem.
100. Mack, T. M., Noble, J. Jr. & Thomas, D. B. (2012). 270, 15974–15978 (1995).
A prospective study of serum antibody and protection 123. Kennedy, R. & Poland, G. A. T-cell epitope discovery 147. Xu, R. H. et al. The orthopoxvirus type I IFN binding
against smallpox. Am. J. Trop. Med. Hyg. 21, for variola and vaccinia viruses. Rev. Med. Virol. 17, protein is essential for virulence and an effective target
214–218 (1972). 93–113 (2007). for vaccination. J. Exp. Med. 205, 981–992 (2008).
101. Keasey, S. et al. Proteomic basis of the antibody 124. Sette, A. et al. Definition of epitopes and antigens 148. Fernandez de Marco Mdel, M., Alejo, A., Hudson, P.,
response to monkeypox virus infection examined in recognized by vaccinia specific immune responses: Damon, I. K. & Alcami, A. The highly virulent variola
cynomolgus macaques and a comparison to human their conservation in variola virus sequences, and use and monkeypox viruses express secreted inhibitors of
smallpox vaccination. PLoS ONE 5, e15547 (2011). as a model system to study complex pathogens. Vaccine type I interferon. FASEB J. 24, 1479–1488 (2010).
102. Howard, A. R., Senkevich, T. G. & Moss, B. Vaccinia 27, G21–G26 (2009). 149. Yoshida, M. et al. Local and systemic responses to
virus A26 and A27 proteins form a stable complex 125. Walsh, S. R. et al. Diverse recognition of conserved SARS-CoV-2 infection in children and adults. Nature
tethered to mature virions by association with the A17 orthopoxvirus CD8+ T cell epitopes in vaccinated 602, 321–327 (2022).
transmembrane protein. J. Virol. 82, 12384–12391 rhesus macaques. Vaccine 27, 4990–5000 (2009). 150. Hijano, D. R. et al. Role of type I interferon (IFN) in the
(2008). 126. Calvo-Calle, J. M., Strug, I., Nastke, M. D., Baker, S. P. respiratory syncytial virus (RSV) immune response and
103. Chiu, W. L., Lin, C. L., Yang, M. H., Tzou, D. L. & & Stern, L. J. Human CD4+ T cell epitopes from disease severity. Front. Immunol. 10, 566 (2019).
Chang, W. Vaccinia virus 4c (A26L) protein on vaccinia virus induced by vaccination or infection. 151. Oeckinghaus, A. & Ghosh, S. The NF-κB family of
intracellular mature virus binds to the extracellular PLoS Pathog. 3, 1511–1529 (2007). transcription factors and its regulation. Cold Spring
cellular matrix laminin. J. Virol. 81, 2149–2157 127. Kennedy, R. B. & Poland, G. A. The identification of Harb. Perspect. Biol. 1, a000034 (2009).
(2007). HLA class II-restricted T cell epitopes to vaccinia virus 152. Bloomer, D. T. et al. CrmA orthologs from diverse
104. Mucker, E. M. et al. Intranasal monkeypox marmoset membrane proteins. Virology 408, 232–240 (2010). poxviruses potently inhibit caspases-1 and -8, yet
model: prophylactic antibody treatment provides 128. Molero-Abraham, M., Glutting, J. P., Flower, D. R., cleavage site mutagenesis frequently produces
benefit against severe monkeypox virus disease. Lafuente, E. M. & Reche, P. A. EPIPOX: caspase-1-specific variants. Biochem. J. 476,
PLoS Negl. Trop. Dis. 12, e0006581 (2018). immunoinformatic characterization of the shared 1335–1357 (2019).
This study highlights the feasibility of using T-cell epitome between variola virus and related 153. Komiyama, T. et al. Inhibition of interleukin-1β
nanoparticle technology as a vehicle for delivering pathogenic orthopoxviruses. J. Immunol. Res. 2015, converting enzyme by the cowpox virus serpin CrmA.
antibody-encoding mRNA targeting poxviruses. 738020 (2015). An example of cross-class inhibition. J. Biol. Chem.
105. Wolffe, E. J., Vijaya, S. & Moss, B. A myristylated 129. Ando, J., Ngo, M. C., Ando, M., Leen, A. & 269, 19331–19337 (1994).
membrane protein encoded by the vaccinia virus L1R Rooney, C. M. Identification of protective T-cell 154. Veyer, D. L. et al. Analysis of the anti-apoptotic activity
open reading frame is the target of potent neutralizing antigens for smallpox vaccines. Cytotherapy 22, of four vaccinia virus proteins demonstrates that B13
monoclonal antibodies. Virology 211, 53–63 (1995). 642–652 (2020). is the most potent inhibitor in isolation and during
106. Chen, Z. et al. Chimpanzee/human mAbs to vaccinia In this study, the E3L gene, which is conserved viral infection. J. Gen. Virol. 95, 2757–2768 (2014).
virus B5 protein neutralize vaccinia and smallpox across orthopoxviruses, is identified as a CD8+ 155. Quan, L. T., Caputo, A., Bleackley, R. C., Pickup, D. J.
viruses and protect mice against vaccinia virus. T cell specific epitope, which could be used to & Salvesen, G. S. Granzyme B is inhibited by the
Proc. Natl Acad. Sci. USA 103, 1882–1887 (2006). monitor the protective immunity following infection. cowpox virus serpin cytokine response modifier A.
107. Mucker, E. M., Thiele-Suess, C., Baumhof, P. 130. Arndt, W. D. et al. Evasion of the innate immune type I J. Biol. Chem. 270, 10377–10379 (1995).
& Hooper, J. W. Lipid nanoparticle delivery of interferon system by monkeypox virus. J. Virol. 89, 156. Devadas, S. et al. Granzyme B is critical for T cell
unmodified mRNAs encoding multiple monoclonal 10489–10499 (2015). receptor-induced cell death of type 2 helper T cells.
antibodies targeting poxviruses in rabbits. Mol. Ther. 131. Denzler, K. L. et al. Attenuated NYCBH vaccinia virus Immunity 25, 237–247 (2006).
Nucleic Acids 28, 847–858 (2022). deleted for the E3L gene confers partial protection 157. Alvarez-de Miranda, F. J., Alonso-Sanchez, I.,
108. Gilchuk, I. et al. Cross-neutralizing and protective against lethal monkeypox virus disease in cynomolgus Alcami, A. & Hernaez, B. TNF decoy receptors
human antibody specificities to poxvirus infections. macaques. Vaccine 29, 9684–9690 (2011). encoded by poxviruses. Pathogens 10, 1065 (2021).
Cell 167, 684–694.e9 (2016). 132. Resch, W., Hixson, K. K., Moore, R. J., Lipton, M. S. 158. Hu, F. Q., Smith, C. A. & Pickup, D. J. Cowpox virus
In this study, a panel of mAbs were evaluated for & Moss, B. Protein composition of the vaccinia virus contains two copies of an early gene encoding a
their potential to cross-neutralize orthopoxviruses. mature virion. Virology 358, 233–247 (2007). soluble secreted form of the type II TNF receptor.
109. Karem, K. L. et al. Monkeypox-induced immunity and 133. Nichols, D. B., De Martini, W. & Cottrell, J. Poxviruses Virology 204, 343–356 (1994).
failure of childhood smallpox vaccination to provide utilize multiple strategies to inhibit apoptosis. Viruses 159. Pontejo, S. M., Alejo, A. & Alcami, A. Comparative
complete protection. Clin. Vaccin. Immunol. 14, 9, 215 (2017). biochemical and functional analysis of viral and human
1318–1327 (2007). 134. Shchelkunov, S. N. & Shchelkunova, G. A. Genes that secreted tumor necrosis factor (TNF) decoy receptors.
110. MacLeod, M. K., Clambey, E. T., Kappler, J. W. control vaccinia virus immunogenicity. Acta Nat. 12, J. Biol. Chem. 290, 15973–15984 (2015).
& Marrack, P. CD4 memory T cells: what are they 33–41 (2020). 160. Marotte, H. & Cimaz, R. Etanercept–TNF receptor
and what can they do? Semin. Immunol. 21, 53–61 135. Smith, G. L. et al. Vaccinia virus immune evasion: and IgG1 Fc fusion protein: is it different from other
(2009). mechanisms, virulence and immunogenicity. J. Gen. TNF blockers? Expert. Opin. Biol. Ther. 14, 569–572
111. Jayani, I., Susmiati, E. & Sulistyawati, W. The correlation Virol. 94, 2367–2392 (2013). (2014).
between CD4 count cell and opportunistic infection 136. Yu, H., Bruneau, R. C., Brennan, G. & Rothenburg, S. 161. Bahar, M. W. et al. Structure and function of A41,
among HIV/AIDS patients. J. Phys. Conf. Ser. 1569, Battle royale: innate recognition of poxviruses a vaccinia virus chemokine binding protein.
032066 (2020). and viral immune evasion. Biomedicines 9, 765 PLoS Pathog. 4, e5 (2008).
112. Smith, Y. E. et al. Smallpox vaccine does not protect (2021). 162. Kotwal, G. J. & Moss, B. Vaccinia virus encodes a
macaques with AIDS from a lethal monkeypox virus 137. Franz, K. M. & Kagan, J. C. Innate immune receptors secretory polypeptide structurally related to complement
challenge. J. Infect. Dis. 191, 372–381 (2005). as competitive determinants of cell fate. Mol. Cell 66, control proteins. Nature 335, 176–178 (1988).
113. Sklenovská, N. & Van Ranst, M. Emergence of 750–760 (2017). 163. Miller, C. G., Shchelkunov, S. N. & Kotwal, G. J.
monkeypox as the most important orthopoxvirus 138. Mogensen, T. H. Pathogen recognition and The cowpox virus-encoded homolog of the vaccinia
infection in humans. Front. Public Health 6, 241 inflammatory signaling in innate immune defenses. virus complement control protein is an inflammation
(2018). Clin. Microbiol. Rev. 22, 240–273 (2009). modulatory protein. Virology 229, 126–133 (1997).
114. Dai, R., Huang, X. & Yang, Y. γδT cells are required 139. Di Pilato, M., Mejias-Perez, E., Sorzano, C. O. S. & 164. Ruiz-Arguello, M. B. et al. An ectromelia virus protein
for CD8+ T cell response to vaccinia viral infection. Esteban, M. Distinct roles of vaccinia virus NF-κB that interacts with chemokines through their
Front. Immunol. 12, 727046 (2021). inhibitor proteins A52, B15, and K7 in the immune glycosaminoglycan binding domain. J. Virol. 82,
115. Goulding, J. et al. CD8 T cells use IFN-γ to protect response. J. Virol. 91, e00575-17 (2017). 917–926 (2008).
against the lethal effects of a respiratory poxvirus 140. Graham, S. C. et al. Vaccinia virus proteins A52 165. Seregin, S. V., Babkina, I. N., Nesterov, A. E.,
infection. J. Immunol. 192, 5415 (2014). and B14 share a Bcl-2-like fold but have evolved to Sinyakov, A. N. & Shchelkunov, S. N. Comparative
116. Remakus, S. et al. Memory CD8+ T cells can outsource inhibit NF-κB rather than apoptosis. PLoS Pathog. 4, studies of γ-interferon receptor-like proteins of variola
IFN-γ production but not cytolytic killing for antiviral e1000128 (2008). major and variola minor viruses. FEBS Lett. 382,
protection. Cell Host Microbe 13, 546–557 (2013). 141. Maluquer de Motes, C. et al. Inhibition of apoptosis 79–83 (1996).
117. McClain, D. J. et al. Immunologic responses to and NF-κB activation by vaccinia protein N1 occur via 166. Upton, C., Mossman, K. & McFadden, G. Encoding
vaccinia vaccines administered by different parenteral distinct binding surfaces and make different contributions of a homolog of the IFN-γ receptor by myxoma virus.
routes. J. Infect. Dis. 175, 756–763 (1997). to virulence. PLoS Pathog. 7, e1002430 (2011). Science 258, 1369–1372 (1992).
118. Ennis, F. A., Cruz, J., Demkowicz, W. E. Jr, Rothman, A. L. 142. Chang, H. W., Watson, J. C. & Jacobs, B. L. The E3L 167. Uvarova, E. A. & Shchelkunov, S. N. Species-specific
& McClain, D. J. Primary induction of human CD8+ gene of vaccinia virus encodes an inhibitor of the differences in the structure of orthopoxvirus
cytotoxic T lymphocytes and interferon-γ-producing interferon-induced, double-stranded RNA-dependent complement-binding protein. Virus Res. 81, 39–45
T cells after smallpox vaccination. J. Infect. Dis. 185, protein kinase. Proc. Natl Acad. Sci. USA 89, (2001).
1657–1659 (2002). 4825–4829 (1992). 168. Liszewski, M. K. et al. Structure and regulatory
119. Miller, J. D. et al. Human effector and memory CD8+ 143. Silverman, R. H. Caps off to poxviruses. Cell Host profile of the monkeypox inhibitor of complement:
T cell responses to smallpox and yellow fever vaccines. Microbe 17, 287–289 (2015). comparison to homologs in vaccinia and variola
Immunity 28, 710–722 (2008). 144. Zhang, P., Jacobs, B. L. & Samuel, C. E. Loss of and evidence for dimer formation. J. Immunol. 176,
120. Munier, C. M. L. et al. The primary immune response protein kinase PKR expression in human HeLa cells 3725–3734 (2006).
to vaccinia virus vaccination includes cells with a complements the vaccinia virus E3L deletion mutant 169. Estep, R. D. et al. Deletion of the monkeypox virus
distinct cytotoxic effector CD4 T-cell phenotype. phenotype by restoration of viral protein synthesis. inhibitor of complement enzymes locus impacts the
Vaccine 34, 5251–5261 (2016). J. Virol. 82, 840–848 (2008). adaptive immune response to monkeypox virus in a
0123456789();:
Reviews
nonhuman primate model of infection. J. Virol. 85, 189. Berhanu, A. et al. Treatment with the smallpox 212. Heymann, D. L., Szczeniowski, M. & Esteves, K.
9527–9542 (2011). antiviral tecovirimat (ST-246) alone or in combination Re-emergence of monkeypox in Africa: a review of the
170. Campbell, J. A., Trossman, D. S., Yokoyama, W. M. with ACAM2000 vaccination is effective as a past six years. Br. Med. Bull. 54, 693–702 (1998).
& Carayannopoulos, L. N. Zoonotic orthopoxviruses postsymptomatic therapy for monkeypox virus 213. Breman, J. G. et al. Human monkeypox, 1970–79.
encode a high-affinity antagonist of NKG2D. infection. Antimicrob. Agents Chemother. 59, Bull. World Health Organ. 58, 165–182 (1980).
J. Exp. Med. 204, 1311–1317 (2007). 4296–4300 (2015). 214. Breman, J. G., Nakano, J. H., Coffi, E., Godfrey, H. &
171. Dasgupta, A., Hammarlund, E., Slifka, M. K. & Fruh, K. 190. Huggins, J. et al. Nonhuman primates are protected Gautun, J. C. Human poxvirus disease after smallpox
Cowpox virus evades CTL recognition and inhibits from smallpox virus or monkeypox virus challenges by eradication. Am. J. Trop. Med. Hyg. 26, 273–281
the intracellular transport of MHC class I molecules. the antiviral drug ST-246. Antimicrob. Agents Chemother. (1977).
J. Immunol. 178, 1654–1661 (2007). 53, 2620–2625 (2009). 215. Foster, S. O. et al. Human monkeypox. Bull. World
172. Byun, M. et al. Two mechanistically distinct immune 191. Russo, A. T. et al. Effects of treatment delay on efficacy Health Organ. 46, 569–576 (1972).
evasion proteins of cowpox virus combine to avoid of tecovirimat following lethal aerosol monkeypox 216. Centers for Disease Control and Prevention. Human
antiviral CD8 T cells. Cell Host Microbe 6, 422–432 virus challenge in cynomolgus macaques. J. Infect. Dis. monkeypox — Kasai Oriental, Democratic Republic
(2009). 218, 1490–1499 (2018). of Congo, February 1996–October 1997. MMWR
173. Born, T. L. et al. A poxvirus protein that binds to 192. Hutson, C. L. et al. Pharmacokinetics and efficacy Morb. Mortal. Wkly. Rep. 46, 1168–1171 (1997).
and inactivates IL-18, and inhibits NK cell response. of a potential smallpox therapeutic, brincidofovir, 217. Jezek, Z. et al. Four generations of probable
J. Immunol. 164, 3246–3254 (2000). in a lethal monkeypox virus animal model. mSphere person-to-person transmission of human monkeypox.
174. Petersen, B. W., Harms, T. J., Reynolds, M. G. & 6, e00927-20 (2021). Am. J. Epidemiol. 123, 1004–1012 (1986).
Harrison, L. H. Use of vaccinia virus smallpox vaccine 193. Stabenow, J. et al. A mouse model of lethal infection 218. Jezek, Z., Grab, B., Szczeniowski, M., Paluku, K. M.
in laboratory and health care personnel at risk for for evaluating prophylactics and therapeutics against & Mutombo, M. Clinico-epidemiological features of
occupational exposure to orthopoxviruses — monkeypox virus. J. Virol. 84, 3909–3920 (2010). monkeypox patients with an animal or human source
recommendations of the Advisory Committee on 194. Andrei, G. & Snoeck, R. Cidofovir activity against of infection. Bull. World Health Organ. 66, 459–464
Immunization Practices (ACIP), 2015. MMWR poxvirus infections. Viruses 2, 2803–2830 (2010). (1988).
Morb. Mortal. Wkly. Rep. 65, 257–262 (2016). 195. Lalezari, J. P. et al. (S)-1-[3-hydroxy-2- 219. Khodakevich, L. et al. Monkey pox virus infection
175. Rao, A. K. et al. Use of JYNNEOS (smallpox and (phosphonylmethoxy)propyl]cytosine (cidofovir): in humans in the Central African Republic [French].
monkeypox vaccine, live, nonreplicating) for preexposure results of a phase I/II study of a novel antiviral Bull. Soc. Pathol. Exot. Filiales 78, 311–320 (1985).
vaccination of persons at risk for occupational exposure nucleotide analogue. J. Infect. Dis. 171, 788–796 220. Merouze, F. & Lesoin, J. J. Monkeypox: second human
to orthopoxviruses: recommendations of the Advisory (1995). case observed in Ivory Coast (rural health sector of
Committee on Immunization Practices — United States, 196. Ortiz, A. et al. Tubular cell apoptosis and Daloa) [French]. Med. Trop. 43, 145–147 (1983).
2022. MMWR Morb. Mortal. Wkly. Rep. 71, 734–742 cidofovir-induced acute renal failure. Antivir. Ther. 221. Meyer, A. et al. First appearance of monkey pox in
(2022). 10, 185–190 (2005). human beings in Gabon [French]. Med. Trop. 51,
176. Nalca, A. & Zumbrun, E. E. ACAM2000: the new 197. Wachsman, M. et al. Pharmacokinetics, safety and 53–57 (1991).
smallpox vaccine for United States strategic national bioavailability of HPMPC (cidofovir) in human 222. Tchokoteu, P. F. et al. Variola or a severe case of
stockpile. Drug Des. Devel. Ther. 4, 71–79 (2010). immunodeficiency virus-infected subjects. Antivir. Res. varicella? A case of human variola due to monkeypox
177. Sukhumvittaya, S., Ampol, S., Pattanapanyasat, K. 29, 153–161 (1996). virus in a child from the Cameroon [French]. Ann. Soc.
& Kantakamalakul, W. Polyfunctional T cell and 198. Hopkins, R. J. & Lane, J. M. Clinical efficacy Belg. Med. Trop. 71, 123–128 (1991).
neutralizing antibody responses to ACAM2000TM of intramuscular vaccinia immune globulin: 223. Doshi, R. H. et al. Epidemiologic and ecologic
smallpox vaccine immunization in primary-vaccinated a literature review. Clin. Infect. Dis. 39, 819–826 investigations of monkeypox, Likouala department,
individuals. Adv. Microbiol. 6, 169–177 (2016). (2004). Republic of the Congo, 2017. Emerg. Infect. Dis. 25,
178. Precopio, M. L. et al. Immunization with vaccinia virus 199. Dubois, M. E., Hammarlund, E. & Slifka, M. K. 281–289 (2019).
induces polyfunctional and phenotypically distinctive Optimization of peptide-based ELISA for serological 224. Reynolds, M. G. et al. Detection of human monkeypox
CD8+ T cell responses. J. Exp. Med. 204, 1405–1416 diagnostics: a retrospective study of human in the Republic of the Congo following intensive
(2007). monkeypox infection. Vector Borne Zoonotic Dis. 12, community education. Am. J. Trop. Med. Hyg. 88,
179. Pugh, C. et al. Human antibody responses to the 400–409 (2012). 982–985 (2013).
polyclonal Dryvax vaccine for smallpox prevention can 200. Ebola ça Suffit Ring Vaccination Trial Consortium. 225. Formenty, P. et al. Human monkeypox outbreak
be distinguished from responses to the monoclonal The ring vaccination trial: a novel cluster randomised caused by novel virus belonging to Congo Basin clade,
replacement vaccine ACAM2000. Clin. Vaccin. Immunol. controlled trial design to evaluate vaccine efficacy and Sudan, 2005. Emerg. Infect. Dis. 16, 1539–1545
21, 877–885 (2014). effectiveness during outbreaks, with special reference (2010).
180. Keckler, M. S., Reynolds, M. G., Damon, I. K. & to Ebola. BMJ 351, h3740 (2015). 226. Reynolds, M. G. et al. Human monkeypox in Sierra
Karem, K. L. The effects of post-exposure smallpox 201. Heinonen, S. et al. Immune profiles provide insights Leone after 44-year absence of reported cases.
vaccination on clinical disease presentation: addressing into respiratory syncytial virus disease severity in Emerg. Infect. Dis. 25, 1023–1025 (2019).
the data gaps between historical epidemiology and young children. Sci. Transl. Med. 12, eaaw0268 227. Berthet, N. et al. Maculopapular lesions in the Central
modern surrogate model data. Vaccine 31, 5192–5201 (2020). African Republic. Lancet 378, 1354 (2011).
(2013). 202. Toh, Z. Q. et al. Comparison of seroconversion in 228. Nakoune, E. et al. A nosocomial outbreak of
181. Keckler, M. S. et al. IMVAMUNE® and ACAM2000® children and adults with mild COVID-19. JAMA Netw. human monkeypox in the Central African Republic.
provide different protection against disease Open 5, e221313 (2022). Open Forum Infect. Dis. 4, ofx168 (2017).
when administered postexposure in an intranasal 203. Cohen, C. A. et al. SARS-CoV-2 specific T cell 229. Yinka-Ogunleye, A. et al. Outbreak of human
monkeypox challenge prairie dog model. Vaccines 8, responses are lower in children and increase with age monkeypox in Nigeria in 2017–18: a clinical and
396 (2020). and time after infection. Nat. Commun. 12, 4678 epidemiological report. Lancet Infect. Dis. 19,
This study evaluates and characterizes the (2021). 872–879 (2019).
differences in protection provided by IMVAMUNE 204. Ryan, M. A. & Seward, J. F., Smallpox Vaccine in 230. Reynolds, M. G. et al. Spectrum of infection and risk
and ACAM2000 as post-exposure prophylactic Pregnancy Registry Team. Pregnancy, birth, and infant factors for human monkeypox, United States, 2003.
vaccines in protection against MPXV infection. health outcomes from the national smallpox vaccine Emerg. Infect. Dis. 13, 1332–1339 (2007).
182. Luong Nguyen, L. B. et al. A prospective national in pregnancy registry, 2003–2006. Clin. Infect. Dis. 231. Erez, N. et al. Diagnosis of imported monkeypox,
cohort evaluating ring MVA vaccination as post-exposure 46, S221–S226 (2008). Israel, 2018. Emerg. Infect. Dis. 25, 980–983 (2019).
prophylaxis for monkeypox. Nat. Med. https://doi.org/ 205. UK Health Security Agency. Monkeypox outbreak: 232. Vaughan, A. et al. Two cases of monkeypox imported
10.1038/d41591-022-00077-1 (2022). vaccination strategy. gov.uk https://www.gov.uk/ to the United Kingdom, September 2018.
This study highlights the potential usefulness guidance/monkeypox-outbreak-vaccination-strategy Eurosurveillance 23, 1800509 (2018).
of a post-exposure prophylaxis ring vaccination (2022). 233. Vaughan, A. et al. Human-to-human transmission of
as an effort to control spread of MPXV infection 206. Nkengasong, J. N. & Tessema, S. K. Africa needs a monkeypox virus, United Kingdom, October 2018.
in a recruited cohort from France. new public health order to tackle infectious disease Emerg. Infect. Dis. 26, 782–785 (2020).
183. Yang, S. & Yonts, A. BLA clinical review memorandum. threats. Cell 183, 296–300 (2020). 234. Hobson, G. et al. Family cluster of three cases of
FDA https://www.fda.gov/media/131870/download 207. Teo, T. H. et al. Co-infection with chikungunya virus monkeypox imported from Nigeria to the United
(2019). alters trafficking of pathogenic CD8+ T cells into Kingdom, May 2021. Eurosurveillance 26, 2100745
184. Pollard, A. J. & Bijker, E. M. A guide to vaccinology: the brain and prevents Plasmodium-induced (2021).
from basic principles to new developments. Nat. Rev. neuropathology. EMBO Mol. Med. 10, 121–138 235. Yong, S. E. F. et al. Imported monkeypox, Singapore.
Immunol. 21, 83–100 (2021). (2018). Emerg. Infect. Dis. 26, 1826–1830 (2020).
185. Lavelle, E. C. & Ward, R. W. Mucosal vaccines — 208. Teo, T. H. et al. Plasmodium co-infection protects 236. Costello, V. et al. Imported monkeypox from
fortifying the frontiers. Nat. Rev. Immunol. 22, against chikungunya virus-induced pathologies. international traveler, Maryland, USA, 2021.
236–250 (2022). Nat. Commun. 9, 3905 (2018). Emerg. Infect. Dis. 28, 1002–1005 (2022).
186. Qin, L. & Evans, D. H. Genome scale patterns of 209. Torres-Ruesta, A. et al. Malaria abrogates 237. Rao, A. K. et al. Monkeypox in a traveler returning
recombination between coinfecting vaccinia viruses. O’nyong-nyong virus pathologies by restricting virus from Nigeria — Dallas, Texas, July 2021. MMWR
J. Virol. 88, 5277–5286 (2014). infection in nonimmune cells. Life Sci. Alliance 5, Morb. Mortal. Wkly. Rep. 71, 509–516 (2022).
187. Midgley, C. M., Putz, M. M., Weber, J. N. & e202101272 (2022). 238. Zumla, A. et al. Monkeypox outbreaks outside
Smith, G. L. Vaccinia virus strain NYVAC induces 210. Yohannes, E. & Chance, M. R. in Encyclopedia of endemic regions: scientific and social priorities.
substantially lower and qualitatively different human Systems Biology (eds Dubitzky, W., Wolkenhauer, O., Lancet Infect. Dis. https://doi.org/10.1016/
antibody responses compared with strains Lister and Cho, K. H. & Yokota, H.) 2078–2084 (Springer, S1473-3099(22)00354-1 (2022).
Dryvax. J. Gen. Virol. 89, 2992–2997 (2008). 2013). 239. Nigeria Centre for Disease Control. Update
188. Russo, A. T. et al. An overview of tecovirimat for 211. Mulkey, S. B. et al. Neurodevelopmental abnormalities on monkeypox (MPX) in Nigeria. NCDC https://
smallpox treatment and expanded anti-orthopoxvirus in children with in utero Zika virus exposure without ncdc.gov.ng/themes/common/files/sitreps/
applications. Expert. Rev. Anti Infect. Ther. 19, congenital Zika syndrome. JAMA Pediatr. 174, 4f9bc59b967d1f4b19d61358595d3546.pdf
331–344 (2021). 269–276 (2020). (2022).
0123456789();:
Reviews
240. Kraemer, M. U. G. et al. Tracking the 2022 protective immunity against monkeypox. Nat. Med. by core funds at the A*STAR Infectious Diseases Labs
monkeypox outbreak with epidemiological data in 11, 1005–1011 (2005). (ID Labs) through the Biomedical Medical Research Council
real-time. Lancet Infect. Dis. https://doi.org/10.1016/ 246. Sanjuan, R. & Domingo-Calap, P. Mechanisms (BMRC), A*STAR and the PREPARE (Programme for
S1473-3099(22)00359-0 (2022). of viral mutation. Cell Mol. Life Sci. 73, 4433–4448 Research in Epidemic Preparedness and REsponse) Outbreak
This is the first paper that creates an open-access (2016). Research Strategic Funds (PREPARE-SF-2022-001).
database and visualization tool for tracking the 247. Elde, N. C. et al. Poxviruses deploy genomic
occurrences of MPXV infection globally. accordions to adapt rapidly against host antiviral Author contributions
241. Channel News Asia. Singapore confirms imported case defenses. Cell 150, 831–841 (2012). All authors contributed equally to all aspects of the article.
of monkeypox after flight attendant develops fever This paper describes the possibility of poxviruses
and rashes. CNA https://www.channelnewsasia.com/ using genomic accordions as a mechanism of Competing interest
singapore/monkeypox-singapore-imported-case-flight- evading host antiviral responses. The authors declare no competing interests.
attendant-fever-rashes-moh-2760996 (2022). 248. Daskalakis, D., McClung, R. P., Mena, L. & Mermin, J.
242. Isidro, J. et al. Phylogenomic characterization and Monkeypox: avoiding the mistakes of past infectious Peer review information
signs of microevolution in the 2022 multi-country disease epidemics. Ann. Intern. Med. https://doi. Nature Reviews Immunology thanks Jacqueline Williams who
outbreak of monkeypox virus. Nat. Med. https:// org/10.7326/M22-1748 (2022). co-reviewed with Bertram Jacobs, Sathesh Panayampalli and
doi.org/10.1038/s41591-022-01907-y (2022). 249. Centers for Disease Control and Prevention. the other, anonymous, reviewer(s) for their contribution to the
This recent article characterizes the MPXV outbreak Monkeypox and smallpox vaccine guidance. CDC peer review of this work.
genome sequence and highlights the involvement of https://www.cdc.gov/poxvirus/monkeypox/clinicians/
host APOBEC3 protein in the virus evolution. smallpox-vaccine.html (2022). Publisher’s note
243. Luna, N. et al. Phylogenomic analysis of the 250. Kimball, S. U.S. has more than 36,000 Jynneos Springer Nature remains neutral with regard to jurisdictional
monkeypox virus (MPXV) 2022 outbreak: emergence monkeypox vaccine doses immediately available in claims in published maps and institutional affiliations.
of a novel viral lineage. Travel. Med. Infect. Dis. https:// national stockpile. CNBC https://www.cnbc.com/2022/
doi.org/10.1016/j.tmaid.2022.102402 (2022). 06/06/us-has-36000-doses-of-jynneos-monkeypox- Springer Nature or its licensor holds exclusive rights to this
244. Gigante, C. M. et al. Multiple lineages of monkeypox vaccine-immediately-available-in-national-stockpile. article under a publishing agreement with the author(s) or
virus detected in the United States, 2021–2022. html (2022). other rightsholder(s); author self-archiving of the accepted
Preprint at bioRxiv https://doi.org/10.1101/2022. manuscript version of this article is solely governed by the
06.10.495526 (2022). Acknowledgements terms of such publishing agreement and applicable law.
245. Hammarlund, E. et al. Multiple diagnostic techniques The authors thank D. Ackerman of Insight Editing London for
identify previously vaccinated individuals with his valuable inputs on an early draft. This work was supported © Springer Nature Limited 2022
0123456789();: