Last data update: Mar 21, 2025. (Total: 48935 publications since 2009)
Records 1-30 (of 50 Records) |
Query Trace: Hutson CL[original query] |
---|
First clade Ib monkeypox virus infection reported in the Americas - California, November 2024
Levy V , Branzuela A , Hsieh K , Getabecha S , Berumen R 3rd , Saadeh K , Snyder RE , Marek G , Dodson D , Newman A , Hacker JK , Kath C , Minhaj FS , Gigante CM , Gearhart S , Kallen A , Hutson CL , Jacobson K . MMWR Morb Mortal Wkly Rep 2025 74 (4) 44-49 ![]() ![]() A clade I monkeypox virus (MPXV) outbreak is ongoing in the Democratic Republic of the Congo; travel-associated clade I MPXV infections have been reported in non-African countries. In November 2024, San Mateo County Health in California identified an electronic laboratory report of polymerase chain reaction results suggestive of clade I MPXV infection in a male traveler who had recently returned from East Africa. After conferring with the California Department of Public Health (CDPH), a county health department worker visited the patient that same day at his home and obtained skin pustule swab specimens for expedited clade I MPXV testing. Clade I MPXV was confirmed the following day by the CDPH Viral and Rickettsial Disease Laboratory. This was the first reported clade I MPXV infection in the Americas. Among 83 identified contacts, five received JYNNEOS vaccine as postexposure prophylaxis. All contacts were monitored for 21 days; no secondary cases were identified. Patients with mpox-compatible lesions or clinical features should receive MPXV testing, and health care providers should immediately notify public health authorities of suspected clade I MPXV infections (e.g., mpox manifestations and travel history to an area with ongoing clade I MPXV transmission) or upon receiving a nonvariola orthopoxvirus DNA detected, clade II MPXV DNA undetectable test result to trigger additional testing and facilitate the rapid implementation of transmission-based precautions and other preventive public health interventions. |
The rise of mpox in a post-smallpox world
McQuiston JH , McCollum A , Christie A , Torres F , Mermin J , Jernigan DB , Hutson CL . Emerg Infect Dis 2025 31 (1) 27-31 ![]() Reports of mpox are rising in Africa where the disease is endemic and in new countries where the disease has not been previously seen. The 2022 global outbreak of clade II mpox and an ongoing outbreak of the more lethal clade I mpox highlight the pandemic potential for monkeypox virus. Waning population immunity after the cessation of routine immunization for smallpox plays a key role in the changing epidemiologic patterns of mpox. Sustained human-to-human transmission of mpox is occurring widely in the context of insufficient population immunity, fueling genetic mutations that affect the accuracy of some diagnostic tests and that could lead to changing virulence. Additional research should address complex challenges for control of mpox, including improved diagnostics and medical countermeasures. The availability of vaccines should be expanded not only for outbreak response but also for broader routine use for persons in mpox-endemic countries. |
Bioluminescence imaging to study recombinant orthopoxvirus infection in animal models
Kling C , Smith TG , Baird N , Hutson CL . Methods Mol Biol 2025 2860 175-189 ![]() Bioluminescent images of viral replication in live animals (in vivo) reveal disease dynamics and effects of medical countermeasures over time. After selecting an appropriate orthopoxvirus animal model for the study, a recombinant virus with the firefly luciferase gene inserted in the genome is used to infect the animals. On the day of bioluminescent imaging, the substrate, D-luciferin, is prepared; animals are sedated and injected with the substrate and IVIS imager is utilized; various bioluminescent images are acquired; then animals recover and are able to continue in the study. Ex vivo imaging can also be completed after animals are euthanized at experimental endpoint. This approach allows real-time imaging of viral kinetics within an animal, and analysis of images can provide an additional quantitative measure throughout the study. Bioluminescent imaging not only provides scientific benefits but also benefits to animal welfare. For these reasons, bioluminescent imaging should be considered for any in vivo orthopoxvirus study. |
Identification of raccoon rabies virus variant in a stray kitten: the role of veterinary practitioners in detection and reporting of a non-native zoonotic pathogen-Nebraska, 2023
Carpenter A , Price ER , Stein SR , Beron AJ , Divis A , Mix S , Hess AR , Nelson KM , Wetzel CT , Fredrick J , Huse L , Horn A , Loy DS , Loy JD , Morgan CN , Rodriguez SE , Shelus V , Gigante CM , Hutson CL , Orciari LA , Swedberg C , Boutelle C , Chipman RB , Donahue M , Wallace RM , Buss BF . J Am Vet Med Assoc 2024 1-4 ![]() Rabies is a fatal encephalitic disease affecting all mammals. This report describes identification of raccoon rabies virus variant isolated from a stray kitten in an urban Midwestern city that is nonendemic for this virus variant. The kitten originally presented with nonspecific neurologic abnormalities. Astute veterinary, wildlife, and public health professionals played a critical role in the identification of this fatal zoonotic disease and the extensive public health and wildlife management response that ensued. This case serves as an important reminder of the risk of rabies in unvaccinated animals or those without known vaccination status, including stray animals. |
Successful collaborations that resulted in increased U.S. diagnostic testing during the 2022 Mpox outbreak
Hutson CL , Villanueva J , Stenzel T , Olson VA , Gerald N , McNall R , Courtney S , Aden T , Rager S , Egan C , Blevins P , Kuhnert W , Davidson W , Khan T , Baird N , Kling C , Van Meter S , Chaitram J , Salerno RM . J Public Health Manag Pract 2024 CONTEXT: The first case of mpox was detected in the United States in a Laboratory Response Network (LRN) laboratory at the Massachusetts Department of Public Health on May 17, 2022. Through previous years of smallpox preparedness efforts by the United States government, testing capacity in LRN laboratories across the United States utilizing the FDA-cleared Centers for Disease Control and Prevention (CDC) Non-variola orthopoxvirus (NVO) test was approximately 6000 tests weekly across the nation prior to the mpox outbreak. By early June 2022, the LRN laboratories had capacity to perform up to 8000 tests per week. As the outbreak expanded, cases were identified in every United States state, peaking at ~3000 cases per week nationally in August 2022. OBJECTIVE: Although NVO testing capacity in LRN laboratories exceeded national mpox testing demand overall, LRN testing access in some areas was challenged and test expansion was necessary. PARTICIPANTS: CDC engaged with partners and select commercial laboratories early to increase diagnostic testing access by allowing these commercial laboratories to utilize the NVO test. SETTING: The expansion of testing to commercial laboratories increased testing availability, capacity, and volume nationwide. This was the first time that CDC shared an FDA 510k-cleared molecular test with commercial laboratories to support a public health emergency. DESIGN: Extensive efforts were made to ensure the CDC NVO test was used appropriately in the private sector and that the transfer process met regulatory requirements. MAIN OUTCOME MEASURES, RESULTS, CONCLUSIONS: These novel methods to expand NVO testing to commercial laboratories increased national testing capacity to 80 000 mpox tests/week. Test volumes among these laboratories never exceeded this expanded capacity. The rapid increase in the nation's testing capacity, in conjunction and coordination with other public and private health efforts, helped to detect cases rapidly. These actions demonstrated the importance of highly functional and efficient public health and private sector partnerships for responding to public health emergencies. |
Notes from the field: Enhanced surveillance for raccoon rabies virus variant and vaccination of wildlife for management - Omaha, Nebraska, October 2023-July 2024
Stein SR , Beron AJ , Nelson KM , Price E , Rodriguez SE , Shelus V , Carpenter A , Hess AR , Boutelle C , Morgan CN , Gigante CM , Hutson CL , Loy JD , Loy DS , Wetzel C , Frederick J , Huse L , Orciari L , Chipman RB , Wallace RM , Donahue M , Buss BF . MMWR Morb Mortal Wkly Rep 2024 73 (41) 933-935 ![]() On September 28, 2023, a kitten aged approximately 6 weeks found in Omaha, Nebraska, had test results positive for rabies at the Nebraska Veterinary Diagnostic Center (NVDC) after dying with neurologic signs and having bitten and scratched its caretakers. Preliminary investigation identified 10 exposed persons for whom postexposure prophylaxis (PEP)(†) was recommended. Subsequent variant-typing by NVDC yielded a presumptive positive result for the Eastern raccoon rabies virus variant (RRVV), which CDC confirmed on October 6. |
Notes from the field: Mpox cluster caused by tecovirimat-resistant monkeypox virus - Five States, October 2023-February 2024
Gigante CM , Takakuwa J , McGrath D , Kling C , Smith TG , Peng M , Wilkins K , Garrigues JM , Holly T , Barbian H , Kittner A , Haydel D , Ortega E , Richardson G , Hand J , Hacker JK , Espinosa A , Haw M , Kath C , Bielby M , Short K , Johnson K , De La Cruz N , Davidson W , Hughes C , Green NM , Baird N , Rao AK , Hutson CL . MMWR Morb Mortal Wkly Rep 2024 73 (40) 903-905 ![]() ![]() The antiviral drug tecovirimat* has been used extensively to treat U.S. mpox cases since the start of a global outbreak in 2022. Mutations in the mpox viral protein target (F13 or VP37) that occur during treatment can result in resistance to tecovirimat(†) (1,2). CDC and public health partners have conducted genetic surveillance of monkeypox virus (MPXV) for F13 mutations through sequencing and monitoring of public databases. MPXV F13 mutations associated with resistance have been reported since 2022, typically among severely immunocompromised mpox patients who required prolonged courses of tecovirimat (3-5). A majority of patients with infections caused by MPXV with resistant mutations had a history of tecovirimat treatment; however, spread of tecovirimat-resistant MPXV was reported in California during late 2022 to early 2023 among persons with no previous tecovirimat treatment (3). This report describes a second, unrelated cluster of tecovirimat-resistant MPXV among 18 persons with no previous history of tecovirimat treatment in multiple states. |
Investigation of an mpox outbreak affecting many vaccinated persons in Chicago, IL-March 2023-June 2023
Faherty EAG , Holly T , Ogale YP , Spencer H , Becht AM , Crisler G , Wasz M , Stonehouse P , Barbian HJ , Zelinski C , Kittner A , Foulkes D , Anderson KW , Evans T , Nicolae L , Staton A , Hardnett C , Townsend MB , Carson WC , Satheshkumar PS , Hutson CL , Gigante CM , Quilter LAS , Gorman S , Borah B , Black SR , Pacilli M , Kern D , Kerins J , McCollum AM , Rao AK , Tabidze I . Clin Infect Dis 2024 79 (1) 122-129 ![]() ![]() BACKGROUND: After months of few mpox cases, an increase in cases was reported in Chicago during May 2023, predominantly among fully vaccinated (FV) patients. We investigated the outbreak scope, differences between vaccinated and unvaccinated patients, and hypotheses for monkeypox virus (MPXV) infection after vaccination. METHODS: We interviewed patients and reviewed medical records to assess demographic, behavioral, and clinical characteristics; mpox vaccine status; and vaccine administration routes. We evaluated serum antibody levels after infection and compared patient viral genomes with MPXV sequences in available databases. We discussed potential vaccine compromise with partners who manufactured, handled, and administered the vaccine associated with breakthrough infections. RESULTS: During 18 March-27 June 2023, we identified 49 mpox cases; 57% of these mpox patients were FV. FV patients received both JYNNEOS doses subcutaneously (57%), intradermally (7%), or via heterologous administration (36%). FV patients had more median sex partners (3; interquartile range [IQR] = 1-4) versus not fully vaccinated patients (1; IQR = 1-2). Thirty-six of 37 sequenced specimens belonged to lineage B.1.20 of clade IIb MPXV, which did not demonstrate any amino acid changes relative to B.1, the predominant lineage from May 2022. Vaccinated patients demonstrated expected humoral antibody responses; none were hospitalized. No vaccine storage excursions were identified. Approximately 63% of people at risk for mpox in Chicago were FV during this period. CONCLUSIONS: Our investigation indicated that cases were likely due to frequent behaviors associated with mpox transmission, even with relatively high vaccine effectiveness and vaccine coverage. Cases after vaccination might occur in similar populations. |
Notes from the field: Clade II mpox surveillance update - United States, October 2023-April 2024
Tuttle A , Hughes CM , Dvorak M , Aeschleman L , Davidson W , Wilkins K , Gigante C , Satheshkumar PS , Rao AK , Minhaj FS , Christensen BE , McQuiston JH , Hutson CL , McCollum AM . MMWR Morb Mortal Wkly Rep 2024 73 (20) 474-476 |
U.S. preparedness and response to increasing clade I mpox cases in the Democratic Republic of the Congo - United States, 2024
McQuiston JH , Luce R , Kazadi DM , Bwangandu CN , Mbala-Kingebeni P , Anderson M , Prasher JM , Williams IT , Phan A , Shelus V , Bratcher A , Soke GN , Fonjungo PN , Kabamba J , McCollum AM , Perry R , Rao AK , Doty J , Christensen B , Fuller JA , Baird N , Chaitram J , Brown CK , Kirby AE , Fitter D , Folster JM , Dualeh M , Hartman R , Bart SM , Hughes CM , Nakazawa Y , Sims E , Christie A , Hutson CL . MMWR Morb Mortal Wkly Rep 2024 73 (19) 435-440 ![]() ![]() Clade I monkeypox virus (MPXV), which can cause severe illness in more people than clade II MPXVs, is endemic in the Democratic Republic of the Congo (DRC), but the country has experienced an increase in suspected cases during 2023-2024. In light of the 2022 global outbreak of clade II mpox, the increase in suspected clade I cases in DRC raises concerns that the virus could spread to other countries and underscores the importance of coordinated, urgent global action to support DRC's efforts to contain the virus. To date, no cases of clade I mpox have been detected outside of countries in Central Africa where the virus is endemic. CDC and other partners are working to support DRC's response. In addition, CDC is enhancing U.S. preparedness by raising awareness, strengthening surveillance, expanding diagnostic testing capacity for clade I MPXV, ensuring appropriate specimen handling and waste management, emphasizing the importance of appropriate medical treatment, and communicating guidance on the recommended contact tracing, containment, behavior modification, and vaccination strategies. |
Serologic responses to the MVA-based JYNNEOS mpox vaccine in a cohort of participants from the District of Columbia (D.C.)
Griffin I , Berry I , Navarra T , Priyamvada L , Carson WC , Noiman A , Jackson DA , Waltenburg MA , Still W , Lujan L , Beverly J , Willut C , Lee M , Mangla A , Shelus V , Hutson CL , Townsend MB , Satheshkumar PS . Vaccine 2024 We assessed early antibody responses after two doses of JYNNEOS (IMVANEX) mpox vaccine in the District of Columbia (D.C.) in persons at high risk for mpox without characteristic lesions or rash. Participants with PCR mpox negative specimens (oral swab, blood, and/or rectal swab) on the day of receipt of the first vaccine dose and who provided a baseline (day 0) serum sample and at least one serum sample at ∼28, ∼42-56 days, or 180 days post vaccination were included in this analysis. Orthopoxvirus (OPXV)-specific IgG and IgM ELISAs and neutralizing antibody titers were performed, and longitudinal serologic responses were examined. Based on participants' IgG and IgM antibody levels at baseline, they were categorized as naïve or non-naïve. Linear mixed effects regression models were conducted to determine if IgG antibody response over time varied by age, sex, HIV status, and route of administration for both naïve and non-naïve participants. Among both naïve and non-naïve participants IgG seropositivity rates increased until day 42-56, with 89.4 % of naïve and 92.1 % of non-naïve participants having detectable IgG antibodies. The proportion of naive participants with detectable IgG antibodies declined by day 180 (67.7 %) but remained high among non-naïve participants (94.4 %). Neutralizing antibody titers displayed a similar pattern, increasing initially post vaccination but declining by day 180 among naïve participants. There were no significant serologic response differences by age, sex, or HIV status. Serologic response did vary by route of vaccine administration, with those receiving a combination of intradermal and subcutaneous doses displaying significantly higher IgG values than those receiving both doses intradermally. These analyses provide initial insights into the immunogenicity of a two-dose JYNNEOS PEP regimen in individuals at high risk of mpox exposure in the United States. |
Ten years of high-consequence pathogens-research gains, readiness gaps, and future goals
McQuiston JH , Montgomery JM , Hutson CL . Emerg Infect Dis 2024 30 (4) 800-802 ![]() ![]() |
Pathology and monkeypox virus localization in tissues from immunocompromised patients with severe or fatal mpox
Ritter JM , Martines RB , Bhatnagar J , Rao AK , Villalba JA , Silva-Flannery L , Lee E , Bullock HA , Hutson CL , Cederroth T , Harris CK , Hord K , Xu Y , Brown CA , Guccione JP , Miller M , Paddock CD , Reagan-Steiner S . J Infect Dis 2024 ![]() ![]() BACKGROUND: Pathology and monkeypox virus (MPXV) tissue tropism in severe and fatal human mpox is not thoroughly described but can help elucidate the disease pathogenesis and the role of coinfections in immunocompromised patients. METHODS: We analyzed biopsy and autopsy tissues from 22 patients with severe or fatal outcomes to characterize pathology and viral antigen and DNA distribution in tissues by immunohistochemistry and in situ hybridization. Tissue-based testing for coinfections was also performed. RESULTS: Mucocutaneous lesions showed necrotizing and proliferative epithelial changes. Deceased patients with autopsy tissues evaluated had digestive tract lesions, and half had systemic tissue necrosis with thrombotic vasculopathy in lymphoid tissues, lung, or other solid organs. Half also had bronchopneumonia, and one-third had acute lung injury. All cases had MPXV antigen and DNA detected in tissues. Coinfections were identified in 5/16 (31%) biopsy and 4/6 (67%) autopsy cases. DISCUSSION: Severe mpox in immunocompromised patients is characterized by extensive viral infection of tissues and viremic dissemination that can progress despite available therapeutics. Digestive tract and lung involvement are common and associated with prominent histopathological and clinical manifestations. Coinfections may complicate mpox diagnosis and treatment. Significant viral DNA (likely correlating to infectious virus) in tissues necessitates enhanced biosafety measures in healthcare and autopsy settings. |
Tecovirimat resistance in Mpox patients, United States, 2022-2023
Smith TG , Gigante CM , Wynn NT , Matheny A , Davidson W , Yang Y , Condori RE , O'Connell K , Kovar L , Williams TL , Yu YC , Petersen BW , Baird N , Lowe D , Li Y , Satheshkumar PS , Hutson CL . Emerg Infect Dis 2023 29 (12) 2426-2432 ![]() ![]() During the 2022 multinational outbreak of monkeypox virus (MPXV) infection, the antiviral drug tecovirimat (TPOXX; SIGA Technologies, Inc., https://www.siga.com) was deployed in the United States on a large scale for the first time. The MPXV F13L gene homologue encodes the target of tecovirimat, and single amino acid changes in F13 are known to cause resistance to tecovirimat. Genomic sequencing identified 11 mutations previously reported to cause resistance, along with 13 novel mutations. Resistant phenotype was determined using a viral cytopathic effect assay. We tested 124 isolates from 68 patients; 96 isolates from 46 patients were found to have a resistant phenotype. Most resistant isolates were associated with severely immunocompromised mpox patients on multiple courses of tecovirimat treatment, whereas most isolates identified by routine surveillance of patients not treated with tecovirimat remained sensitive. The frequency of resistant viruses remains relatively low (<1%) compared with the total number of patients treated with tecovirimat. |
Prevalence of undiagnosed monkeypox virus infections during global mpox outbreak, United States, June-September 2022
Minhaj FS , Singh V , Cohen SE , Townsend M , Scott H , Szumowski J , Hare CB , Upadhyay P , Reddy J , Alexander B , Baird N , Navarra T , Priyamvada L , Wynn N , Carson WC , Odafe S , Guagliardo SAJ , Sims E , Rao AK , Satheshkumar PS , Weidle PJ , Hutson CL . Emerg Infect Dis 2023 29 (11) 2307-2314 ![]() Since May 2022, mpox has been identified in 108 countries without endemic disease; most cases have been in gay, bisexual, or other men who have sex with men. To determine number of missed cases, we conducted 2 studies during June-September 2022: a prospective serologic survey detecting orthopoxvirus antibodies among men who have sex with men in San Francisco, California, and a retrospective monkeypox virus PCR testing of swab specimens submitted for other infectious disease testing among all patients across the United States. The serosurvey of 225 participants (median age 34 years) detected 18 (8.0%) who were orthopoxvirus IgG positive and 3 (1.3%) who were also orthopoxvirus IgM positive. The retrospective PCR study of 1,196 patients (median age 30 years; 54.8% male) detected 67 (5.6%) specimens positive for monkeypox virus. There are likely few undiagnosed cases of mpox in regions where sexual healthcare is accessible and patient and clinician awareness about mpox is increased. |
How the orthodox features of orthopoxviruses led to an unorthodox Mpox outbreak: What we've learned, and what we still need to understand
Brooks JT , Reynolds MG , Torrone E , McCollum A , Spicknall IH , Gigante CM , Li Y , Satheshkumar PS , Quilter LAS , Rao AK , O'Shea J , Guagliardo SAJ , Townsend M , Hutson CL . J Infect Dis 2023 ![]() ![]() Orthopoxviruses are complex, large-genome DNA viruses that have repeatedly confounded expectations in terms of the clinical illness they cause and their patterns of spread. Monkeypox virus (MPXV) was originally characterized during outbreaks among captive primates in the late 1950's. Human disease (mpox) has been observed since the 1970's and inter-human spread has largely been associated with non-sexual, close physical contact in endemic areas of west and central Africa. In May 2022, a focus of Clade IIb MPXV transmission was detected, spreading largely by sexual contact through international networks of gay, bisexual, and other men who have sex with men. Despite decades of preparedness for the potential biothreat risk posed by smallpox, the outbreak grew in both size and geographic scope, testing the strength of smallpox preparedness tools and public health science alike. In this article we consider what was known about mpox prior to the 2022 outbreak, what we have learned about mpox and Clade IIb virus during the outbreak, and what outbreak response actions and continued research are needed to ensure the global public health community is equipped to detect and halt the further spread of this disease threat. We focus on how epidemiologic characterization and investigation together with laboratory studies have advanced our understanding of the transmission and pathogenesis of mpox, and describe what work remains to be done to optimize diagnostics, therapeutics, and vaccines. Persistent health inequities challenge our capacity to fully eliminate circulation of the 2022 outbreak strain of MPXV currently in the United States. |
Community spread of a human monkeypox virus variant with a tecovirimat resistance-associated mutation
Garrigues JM , Hemarajata P , Espinosa A , Hacker JK , Wynn NT , Smith TG , Gigante CM , Davidson W , Vega J , Edmondson H , Karan A , Marutani AN , Kim M , Terashita D , Balter SE , Hutson CL , Green NM . Antimicrob Agents Chemother 2023 67 (11) e0097223 ![]() ![]() Tecovirimat, also known as TPOXX or ST-246, is a drug available for the treatment of mpox. Tecovirimat targets the conserved orthopoxvirus VP37 protein (also known as F13) required for extracellular virus particle generation (1, 2). Multiple VP37 mutations associated with tecovirimat resistance have been reported within the current global mpox outbreak in immunocompromised individuals with advanced HIV infection (3 – 5). In many of these cases, resistance mutation heterogeneity was observed following tecovirimat exposure, suggesting resistance emerged under selective pressure during treatment. | To monitor circulating monkeypox virus (MPXV) within California, a genomic surveillance network was established whereby clinical and commercial laboratories provided positive specimens for whole-genome sequencing using an amplicon-based protocol and subsequent analysis (6 – 9). Through this surveillance, 11 mpox cases were identified in southern California with the same tecovirimat resistance-associated mutation (Table 1): a three-nucleotide deletion in the vaccinia virus Copenhagen F13L gene homolog (OPG057) resulting in asparagine removed from position 267 in the VP37 protein (VP37:N267del) (5) (https://www.fda.gov/emergency-preparedness-and-response/mcm-issues/fda-mpox-response#therapeutics). VP37:N267del was the only tecovirimat resistance-associated mutation detected in identified specimens and had allele frequencies greater than 89% in all instances, suggesting infections may have occurred with predominantly mutant virus. Phenotypic testing in vitro (3 – 5) confirmed tecovirimat resistance in ten identified specimens with EC50 values ranging from 1.488 to 3.977 µM, corresponding to an 85- to 230-fold change compared to wild-type isolates. |
Resistance to anti-orthopoxviral drug tecovirimat (TPOXX) during the 2022 mpox outbreak in the US (preprint)
Smith TG , Gigante CM , Wynn NT , Matheny A , Davidson W , Yang Y , Condori RE , O'Connell K , Kovar L , Williams TL , Yu YC , Petersen BW , Baird N , Lowe D , Li Y , Satheshkumar PS , Hutson CL . medRxiv 2023 18 Background: During the 2022 multinational outbreak of monkeypox virus (MPXV) clade IIb, the antiviral drug tecovirimat (TPOXX) was deployed in the US on a large scale for the first time ever. The MPXV F13L gene homolog encodes the target of tecovirimat, and single amino acid changes in the F13 protein are known to cause resistance to tecovirimat in orthopoxviruses (OPXV). Method(s): Whole genome metagenomic sequencing and amplicon-based sequencing targeting the F13L gene was used to identify nine mutations previously reported to cause resistance in other OPXV along with ten novel mutations that have been identified from the 2022 mpox outbreak. A cytopathic effect assay, previously established at CDC as part of WHO smallpox research, was adapted to MPXV for tecovirimat phenotype testing of virus isolated from mpox patients. Result(s): As of March 2023, in total, 70 isolates from 40 patients were tested, and 50 of these isolates from 26 patients were found to have a resistant phenotype. Most resistant isolates were associated with severely immunocompromised mpox patients on multiple courses of TPOXX treatment; while isolates with F13 mutations identified by routine surveillance of patients not treated with TPOXX have remained sensitive. Conclusion(s): These data indicate that tecovirimat resistance is developing in immunocompromised patients treated with TPOXX and that for isolates that we have analyzed, the frequency of resistant viruses remain relatively low (< 1%) compared to the total number of patients treated with TPOXX. These findings inform our understanding of when tecovirimat resistance is likely to occur and highlight the need for additional OPXV therapeutics. Copyright The copyright holder for this preprint is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. |
Multiple lineages of Monkeypox virus detected in the United States, 2021-2022 (preprint)
Gigante CM , Korber B , Seabolt MH , Wilkins K , Davidson W , Rao AK , Zhao H , Hughes CM , Minhaj F , Waltenburg MA , Theiler J , Smole S , Gallagher GR , Blythe D , Myers R , Schulte J , Stringer J , Lee P , Mendoza RM , Griffin-Thomas LA , Crain J , Murray J , Atkinson A , Gonzalez AH , Nash J , Batra D , Damon I , McQuiston J , Hutson CL , McCollum AM , Li Y . bioRxiv 2022 11 (6619) 560-565 Monkeypox is a viral zoonotic disease endemic in Central and West Africa. In May 2022, dozens of non-endemic countries reported hundreds of monkeypox cases, most with no epidemiological link to Africa. We identified two lineages of Monkeypox virus (MPXV) among nine 2021 and 2022 U.S. monkeypox cases. A 2021 case was highly similar to the 2022 MPXV outbreak variant, suggesting a common ancestor. Analysis of mutations among these two lineages revealed an extreme preference for GA-to-AA mutations indicative of APOBEC3 cytosine deaminase activity that was shared among West African MPXV since 2017 but absent from Congo Basin lineages. Poxviruses are not thought to be subject to APOBEC3 editing; however, these findings suggest APOBEC3 activity has been recurrent and dominant in recent West African MPXV evolution. Copyright The copyright holder for this preprint is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. |
Genomic deletions and rearrangements in monkeypox virus from the 2022 outbreak, USA (preprint)
Gigante CM , Plumb M , Ruprecht A , Zhao H , Wicker V , Wilkins K , Matheny A , Khan T , Davidson W , Sheth M , Burgin A , Burroughs M , Padilla J , Lee JS , Batra D , Hetrick EE , Howard DT , Garfin J , Tate L , Hubsmith SJ , Mendoza RM , Stanek D , Gillani S , Lee M , Mangla A , Blythe D , SierraPatev S , Carpenter-Azevedo K , Huard RC , Gallagher G , Hall J , Ash S , Kovar L , Seabolt MH , Weigand MR , Damon I , Satheshkumar PS , McCollum AM , Hutson CL , Wang X , Li Y . bioRxiv 2022 17 Genomic surveillance of monkeypox virus (MPXV) during the 2022 outbreak has been mainly focused on single nucleotide polymorphism (SNP) changes. DNA viruses, including MPXV, have a lower SNP mutation rate than RNA viruses due to higher fidelity replication machinery. We identified a large genomic rearrangement in a MPXV sequence from a 2022 case in the state of Minnesota (MN), USA, from an abnormal, uneven MPXV read mapping coverage profile in whole-genome sequencing (WGS) data. We further screened WGS data of 206 U.S. MPXV samples and found seven (3.4 percent) sequenced genomes contained similar abnormal read coverage profiles that suggested putative large deletions or genomic rearrangements. Here, we present three MPXV genomes containing deletions ranging from 2.3 to 15 kb and four genomes containing more complex rearrangements. Five genomic changes were each only seen in one sample, but two sequences from linked cases shared an identical 2.3 kb deletion in the 3' terminal region. All samples were positive using VAC1 and Clade II (formerly West African)-specific MPXV diagnostic tests; however, large deletions and genomic rearrangements like the ones reported here have the potential to result in viruses in which the target of a PCR diagnostic test is deleted. The emergence of genomic rearrangements during the outbreak may have public health implications and highlight the importance of continued genomic surveillance. Copyright The copyright holder for this preprint is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. This article is a US Government work. It is not subject to copyright under 17 USC 105 and is also made available for use under a CC0 license. |
Identification of tecovirimat resistance-associated mutations in human monkeypox virus - Los Angeles County
Garrigues JM , Hemarajata P , Karan A , Shah NK , Alarcón J , Marutani AN , Finn L , Smith TG , Gigante CM , Davidson W , Wynn NT , Hutson CL , Kim M , Terashita D , Balter SE , Green NM . Antimicrob Agents Chemother 2023 67 (7) e0056823 ![]() Tecovirimat (also known as TPOXX or ST-246) is a drug available for the treatment of mpox through the Centers for Disease Control and Prevention’s Expanded Access Investigational New Drug “compassionate use” protocol (https://www.cdc.gov/poxvirus/monkeypox/clinicians/Tecovirimat.html). In Los Angeles County, a fatal case of mpox with tecovirimat resistance was previously reported (1). Epidemiologic surveillance in Los Angeles County has since identified additional cases of severe mpox that did not improve after multiple rounds of tecovirimat treatment, including one involving a person who succumbed to infection (Table 1). Consistent with reports describing severe manifestations of mpox within the current global outbreak (1, 2), the identified cases involved host immunodeficiency due to advanced HIV infection. |
The CDC domestic mpox response - United States, 2022-2023
McQuiston JH , Braden CR , Bowen MD , McCollum AM , McDonald R , Carnes N , Carter RJ , Christie A , Doty JB , Ellington S , Fehrenbach SN , Gundlapalli AV , Hutson CL , Kachur RE , Maitland A , Pearson CM , Prejean J , Quilter LAS , Rao AK , Yu Y , Mermin J . MMWR Morb Mortal Wkly Rep 2023 72 (20) 547-552 Monkeypox (mpox) is a serious viral zoonosis endemic in west and central Africa. An unprecedented global outbreak was first detected in May 2022. CDC activated its emergency outbreak response on May 23, 2022, and the outbreak was declared a Public Health Emergency of International Concern on July 23, 2022, by the World Health Organization (WHO),* and a U.S. Public Health Emergency on August 4, 2022, by the U.S. Department of Health and Human Services.(†) A U.S. government response was initiated, and CDC coordinated activities with the White House, the U.S. Department of Health and Human Services, and many other federal, state, and local partners. CDC quickly adapted surveillance systems, diagnostic tests, vaccines, therapeutics, grants, and communication systems originally developed for U.S. smallpox preparedness and other infectious diseases to fit the unique needs of the outbreak. In 1 year, more than 30,000 U.S. mpox cases were reported, more than 140,000 specimens were tested, >1.2 million doses of vaccine were administered, and more than 6,900 patients were treated with tecovirimat, an antiviral medication with activity against orthopoxviruses such as Variola virus and Monkeypox virus. Non-Hispanic Black (Black) and Hispanic or Latino (Hispanic) persons represented 33% and 31% of mpox cases, respectively; 87% of 42 fatal cases occurred in Black persons. Sexual contact among gay, bisexual, and other men who have sex with men (MSM) was rapidly identified as the primary risk for infection, resulting in profound changes in our scientific understanding of mpox clinical presentation, pathogenesis, and transmission dynamics. This report provides an overview of the first year of the response to the U.S. mpox outbreak by CDC, reviews lessons learned to improve response and future readiness, and previews continued mpox response and prevention activities as local viral transmission continues in multiple U.S. jurisdictions (Figure). |
Mpox respiratory transmission: the state of the evidence.
Beeson A , Styczynski A , Hutson CL , Whitehill F , Angelo KM , Minhaj FS , Morgan C , Ciampaglio K , Reynolds MG , McCollum AM , Guagliardo SAJ . Lancet Microbe 2023 4 (4) e277-e283 ![]() ![]() The relative contribution of the respiratory route to transmission of mpox (formerly known as monkeypox) is unclear. We review the evidence for respiratory transmission of monkeypox virus (MPXV), examining key works from animal models, human outbreaks and case reports, and environmental studies. Laboratory experiments have initiated MPXV infection in animals via respiratory routes. Some animal-to-animal respiratory transmission has been shown in controlled studies, and environmental sampling studies have detected airborne MPXV. Reports from real-life outbreaks demonstrate that transmission is associated with close contact, and although it is difficult to infer the route of MPXV acquisition in individual case reports, so far respiratory transmission has not been specifically implicated. Based on the available evidence, the likelihood of human-to-human MPXV respiratory transmission appears to be low; however, studies should continue to assess this possibility. |
Evidence of mpox virus infection among persons without characteristic lesions or rash presenting for first dose of JYNNEOS vaccine-District of Columbia, August 2022
Ogale YP , Baird N , Townsend MB , Berry I , Griffin I , Lee M , Ashley P , Rhodes T , Notigan T , Wynn N , Kling C , Smith T , Priyamvada L , Carson WC , Navarra T , Dawson P , Weidle PJ , Willut C , Mangla AT , Satheshkumar PS , Hutson CL , Jackson DA , Waltenburg MA . Clin Infect Dis 2023 77 (2) 298-302 We assessed mpox virus prevalence in blood, pharyngeal, and rectal specimens among persons without characteristic rash presenting for JYNNEOS vaccine. Our data indicate that the utility of risk-based screening for mpox in persons without skin lesions or rash via pharyngeal swabs, rectal swabs, and/or blood is likely limited. |
An Mpox-related death in the United States
Alarcón J , Kim M , Terashita D , Davar K , Garrigues JM , Guccione JP , Evans MG , Hemarajata P , Wald-Dickler N , Holtom P , Garcia Tome R , Anyanwu L , Shah NK , Miller M , Smith T , Matheny A , Davidson W , Hutson CL , Lucas J , Ukpo OC , Green NM , Balter SE . N Engl J Med 2023 388 (13) 1246-1247 Since May 2022, when the multinational mpox (formerly known as monkeypox) clade IIb virus outbreak was first reported, more than 30,000 cases have been identified in the United States.1 In one study involving more than 1900 patients with mpox, more than 35% of the patients also had human immunodeficiency virus (HIV) infection.2 | | We report a death due to mpox in a patient in the United States. A 33-year-old man with HIV infection (CD4+ T-cell count, <35 per cubic millimeter) and recently treated syphilis became infected with mpox virus (MPXV) (clade IIb). He received two courses of oral tecovirimat (from Aug. 6 through Aug. 20, 2022, and from Aug. 21 through Sept. 4, 2022) and died on hospital day 27. |
Epidemiology of human mpox - worldwide, 2018-2021
McCollum AM , Shelus V , Hill A , Traore T , Onoja B , Nakazawa Y , Doty JB , Yinka-Ogunleye A , Petersen BW , Hutson CL , Lewis R . MMWR Morb Mortal Wkly Rep 2023 72 (3) 68-72 Monkeypox (mpox) is a zoonotic disease caused by Monkeypox virus (MPXV), an Orthopoxvirus; the wild mammalian reservoir species is not known. There are two genetic clades of MPXV: clade I and clade II (historically found in central and west Africa, respectively), with only Cameroon reporting both clades (1). Human cases have historically been reported from 1) mostly rural, forested areas in some central and west African countries; 2) countries reporting cases related to population migration or travel of infected persons; and 3) exposure to imported infected mammals (2). The annual number of cases in Africa has risen since 2014 and cumulatively surpassed reports from the previous 40 years for most countries. This reemergence of mpox might be due to a combination of environmental and ecological changes, animal or human movement, the cessation of routine smallpox vaccination since its eradication in 1980, improvements in disease detection and diagnosis, and genetic changes in the virus (2). This report describes the epidemiology of mpox since 1970 and during 2018-2021, using data from national surveillance programs, World Health Organization (WHO) bulletins, and case reports, and addresses current diagnostic and treatment challenges in countries with endemic disease. During 2018-2021, human cases were recognized and confirmed in six African countries, with most detected in the Democratic Republic of the Congo (DRC) and Nigeria. The reemergence and increase in cases resulted in its being listed in 2019 as a priority disease for immediate and routine reporting through the Integrated Disease Surveillance and Response strategy in the WHO African region.* In eight instances, patients with mpox were identified in four countries outside of Africa after travel from Nigeria. Since 2018, introductory and intermediate training courses on prevention and control of mpox for public health and health care providers have been available online at OpenWHO.(†)(,)(§) The global outbreak that began in May 2022(¶) has further highlighted the need for improvements in laboratory-based surveillance and access to treatments and vaccines to prevent and contain the infection, including in areas of Africa with endemic mpox. |
Multiple lineages of monkeypox virus detected in the United States, 2021-2022.
Gigante CM , Korber B , Seabolt MH , Wilkins K , Davidson W , Rao AK , Zhao H , Smith TG , Hughes CM , Minhaj F , Waltenburg MA , Theiler J , Smole S , Gallagher GR , Blythe D , Myers R , Schulte J , Stringer J , Lee P , Mendoza RM , Griffin-Thomas LA , Crain J , Murray J , Atkinson A , Gonzalez AH , Nash J , Batra D , Damon I , McQuiston J , Hutson CL , McCollum AM , Li Y . Science 2022 378 (6619) eadd4153 ![]() ![]() Monkeypox is a viral zoonotic disease endemic in Central and West Africa. In May 2022, dozens of non-endemic countries reported hundreds of monkeypox cases, most with no epidemiological link to Africa. We identified two lineages of monkeypox virus (MPXV) among two 2021 and seven 2022 U.S. monkeypox cases: the major 2022 outbreak variant, B.1, and a minor contemporaneously sampled variant called A.2. Analyses of mutations among these two variants revealed an extreme preference for GA-to-AA mutations indicative of human APOBEC3 cytosine deaminase activity among Clade IIb MPXV (previously West African, Nigeria) sampled since 2017. Such mutations were not enriched within other MPXV clades. These findings suggest that APOBEC3 editing may be a recurrent and a dominant driver of MPXV evolution within the current outbreak. |
Orthopoxvirus Testing Challenges for Persons in Populations at Low Risk or Without Known Epidemiologic Link to Monkeypox - United States, 2022.
Minhaj FS , Petras JK , Brown JA , Mangla AT , Russo K , Willut C , Lee M , Beverley J , Harold R , Milroy L , Pope B , Gould E , Beeler C , Schneider J , Mostafa HH , Godfred-Cato S , Click ES , Borah BF , Galang RR , Cash-Goldwasser S , Wong JM , McCormick DW , Yu PA , Shelus V , Carpenter A , Schatzman S , Lowe D , Townsend MB , Davidson W , Wynn NT , Satheshkumar PS , O'Connor SM , O'Laughlin K , Rao AK , McCollum AM , Negrón ME , Hutson CL , Salzer JS . MMWR Morb Mortal Wkly Rep 2022 71 (36) 1155-1158 ![]() Since May 2022, approximately 20,000 cases of monkeypox have been identified in the United States, part of a global outbreak occurring in approximately 90 countries and currently affecting primarily gay, bisexual, and other men who have sex with men (MSM) (1). Monkeypox virus (MPXV) spreads from person to person through close, prolonged contact; a small number of cases have occurred in populations who are not MSM (e.g., women and children), and testing is recommended for persons who meet the suspected case definition* (1). CDC previously developed five real-time polymerase chain reaction (PCR) assays for detection of orthopoxviruses from lesion specimens (2,3). CDC was granted 510(k) clearance for the nonvariola-orthopoxvirus (NVO)-specific PCR assay by the Food and Drug Administration. This assay was implemented within the Laboratory Response Network (LRN) in the early 2000s and became critical for early detection of MPXV and implementation of public health action in previous travel-associated cases as well as during the current outbreak (4-7). PCR assays (NVO and other Orthopoxvirus laboratory developed tests [LDT]) represent the primary tool for monkeypox diagnosis. These tests are highly sensitive, and cross-contamination from other MPXV specimens being processed, tested, or both alongside negative specimens can occasionally lead to false-positive results. This report describes three patients who had atypical rashes and no epidemiologic link to a monkeypox case or known risk factors; these persons received diagnoses of monkeypox based on late cycle threshold (Ct) values ≥34, which were false-positive test results. The initial diagnoses were followed by administration of antiviral treatment (i.e., tecovirimat) and JYNNEOS vaccine postexposure prophylaxis (PEP) to patients' close contacts. After receiving subsequent testing, none of the three patients was confirmed to have monkeypox. Knowledge gained from these and other cases resulted in changes to CDC guidance. When testing for monkeypox in specimens from patients without an epidemiologic link or risk factors or who do not meet clinical criteria (or where these are unknown), laboratory scientists should reextract and retest specimens with late Ct values (based on this report, Ct ≥34 is recommended) (8). CDC can be consulted for complex cases including those that appear atypical or questionable cases and can perform additional viral species- and clade-specific PCR testing and antiorthopoxvirus serologic testing. |
Rapid diagnostic testing for response to the monkeypox outbreak - Laboratory Response Network, United States, May 17-June 30, 2022
Aden TA , Blevins P , York SW , Rager S , Balachandran D , Hutson CL , Lowe D , Mangal CN , Wolford T , Matheny A , Davidson W , Wilkins K , Cook R , Roulo RM , White MK , Berman L , Murray J , Laurance J , Francis D , Green NM , Berumen RA3rd , Gonzalez A , Evans S , Hudziec M , Noel D , Adjei M , Hovan G , Lee P , Tate L , Gose RB , Voermans R , Crew J , Adam PR , Haydel D , Lukula S , Matluk N , Shah S , Featherston J , Ware D , Pettit D , McCutchen E , Acheampong E , Buttery E , Gorzalski A , Perry M , Fowler R , Lee RB , Nickla R , Huard R , Moore A , Jones K , Johnson R , Swaney E , Jaramillo J , Reinoso Webb C , Guin B , Yost J , Atkinson A , Griffin-Thomas L , Chenette J , Gant J , Sterkel A , Ghuman HK , Lute J , Smole SC , Arora V , Demontigny CK , Bielby M , Geeter E , Newman KAM , Glazier M , Lutkemeier W , Nelson M , Martinez R , Chaitram J , Honein MA , Villanueva JM . MMWR Morb Mortal Wkly Rep 2022 71 (28) 904-907 As part of public health preparedness for infectious disease threats, CDC collaborates with other U.S. public health officials to ensure that the Laboratory Response Network (LRN) has diagnostic tools to detect Orthopoxviruses, the genus that includes Variola virus, the causative agent of smallpox. LRN is a network of state and local public health, federal, U.S. Department of Defense (DOD), veterinary, food, and environmental testing laboratories. CDC developed, and the Food and Drug Administration (FDA) granted 510(k) clearance* for the Non-variola Orthopoxvirus Real-time PCR Primer and Probe Set (non-variola Orthopoxvirus [NVO] assay), a polymerase chain reaction (PCR) diagnostic test to detect NVO. On May 17, 2022, CDC was contacted by the Massachusetts Department of Public Health (DPH) regarding a suspected case of monkeypox, a disease caused by the Orthopoxvirus Monkeypox virus. Specimens were collected and tested by the Massachusetts DPH public health laboratory with LRN testing capability using the NVO assay. Nationwide, 68 LRN laboratories had capacity to test approximately 8,000 NVO tests per week during June. During May 17-June 30, LRN laboratories tested 2,009 specimens from suspected monkeypox cases. Among those, 730 (36.3%) specimens from 395 patients were positive for NVO. NVO-positive specimens from 159 persons were confirmed by CDC to be monkeypox; final characterization is pending for 236. Prompt identification of persons with infection allowed rapid response to the outbreak, including isolation and treatment of patients, administration of vaccines, and other public health action. To further facilitate access to testing and increase convenience for providers and patients by using existing provider-laboratory relationships, CDC and LRN are supporting five large commercial laboratories with a national footprint (Aegis Science, LabCorp, Mayo Clinic Laboratories, Quest Diagnostics, and Sonic Healthcare) to establish NVO testing capacity of 10,000 specimens per week per laboratory. On July 6, 2022, the first commercial laboratory began accepting specimens for NVO testing based on clinician orders. |
A cocktail of human monoclonal antibodies broadly neutralizes North American rabies virus variants as a promising candidate for rabies post-exposure prophylaxis.
Ejemel M , Smith TG , Greenberg L , Carson WC , Lowe D , Yang Y , Jackson FR , Morgan CN , Martin BE , Kling C , Hutson CL , Gallardo-Romero N , Ellison JA , Moore S , Buzby A , Sullivan-Bolyai J , Klempner M , Wang Y . Sci Rep 2022 12 (1) 9403 ![]() Human rabies remains a globally significant public health problem. Replacement of polyclonal anti-rabies immunoglobulin (RIG), a passive component of rabies post-exposure prophylaxis (PEP), with a monoclonal antibody (MAb), would eliminate the cost and availability constraints associated with RIG. Our team has developed and licensed a human monoclonal antibody RAB1 (Rabishield()), as the replacement for RIG where canine rabies is enzootic. However, for the highly diverse rabies viruses of North America, a cocktail containing two or more MAbs targeting different antigenic sites of the rabies glycoprotein should be included to ensure neutralization of all variants of the virus. In this study, two MAb cocktails, R172 (RAB1-RAB2) and R173 (RAB1-CR57), were identified and evaluated against a broad range of rabies variants from North America. R173 was found to be the most potent cocktail, as it neutralized all the tested North American RABV isolates and demonstrated broad coverage of isolates from both terrestrial and bat species. R173 could be a promising candidate as an alternative or replacement for RIG PEP in North America. |
- Page last reviewed:Feb 1, 2024
- Page last updated:Mar 21, 2025
- Content source:
- Powered by CDC PHGKB Infrastructure