196 results on '"Seiya Yamayoshi"'
Search Results
52. Casirivimab/Imdevimab for Active COVID-19 Pneumonia Which Persisted for Nine Months in a Patient with Follicular Lymphoma during Anti-CD20 Therapy
- Author
-
Hiroyuki Nagai, Makoto Saito, Eisuke Adachi, Yuko Sakai-Tagawa, Seiya Yamayoshi, Maki Kiso, Toyotaka Kawamata, Michiko Koga, Yoshihiro Kawaoka, Takeya Tsutsumi, and Hiroshi Yotsuyanagi
- Subjects
Microbiology (medical) ,Infectious Diseases ,SARS-CoV-2 ,Humans ,General Medicine ,Antibodies, Viral ,Lymphoma, Follicular ,BNT162 Vaccine ,COVID-19 Drug Treatment - Abstract
Immunocompromised patients are more likely to develop severe COVID-19, and exhibit high mortality. It is also hypothesized that chronic infection in these patients can be a risk factor for developing new variants. We describe a patient with prolonged active infection of COVID-19 who became infected during treatment with an anti-CD20 antibody (obinutuzumab) for follicular lymphoma. This patient had persistent RT-PCR positivity and live virus isolation for nine months despite treatment with remdesivir and other potential antiviral therapies. The computed tomography image of the chest showed that the viral pneumonia repeatedly appeared and disappeared in different lobes, as if a new infection had occurred continuously. The patient's SARS-CoV-2 antibody titer was negative throughout the illness, even after two doses of the BNT162b2 mRNA vaccine were administered in the seventh month of infection. A combination of monoclonal antibody therapy against COVID-19 (casirivimab and imdevimab) and antivirals resulted in negative RT-PCR results, and the virus was no longer isolated. The patient was clinically cured. During the 9-month active infection period, no fixed mutations in the spike (S) protein were detected, and the in vitro susceptibility to remdesivir was retained. Therapeutic administration of anti-SARS-CoV-2 monoclonal antibodies is essential in immunocompromised patients. Therefore, measures to prevent resistance against these key drugs are urgently needed.
- Published
- 2022
53. Efficacy of Antiviral Agents against Omicron Subvariants BQ.1.1 and XBB
- Author
-
Masaki Imai, Mutsumi Ito, Maki Kiso, Seiya Yamayoshi, Ryuta Uraki, Shuetsu Fukushi, Shinji Watanabe, Tadaki Suzuki, Ken Maeda, Yuko Sakai-Tagawa, Kiyoko Iwatsuki-Horimoto, Peter J. Halfmann, and Yoshihiro Kawaoka
- Subjects
General Medicine - Published
- 2023
54. Humoral immune evasion of the omicron subvariants BQ.1.1 and XBB
- Author
-
Ryuta Uraki, Mutsumi Ito, Yuri Furusawa, Seiya Yamayoshi, Kiyoko Iwatsuki-Horimoto, Eisuke Adachi, Makoto Saito, Michiko Koga, Takeya Tsutsumi, Shinya Yamamoto, Amato Otani, Maki Kiso, Yuko Sakai-Tagawa, Hiroshi Ueki, Hiroshi Yotsuyanagi, Masaki Imai, and Yoshihiro Kawaoka
- Subjects
Infectious Diseases ,Humans ,Immune Evasion - Published
- 2023
55. Therapeutic efficacy of monoclonal antibodies and antivirals against SARS-CoV-2 Omicron BA.1 in Syrian hamsters
- Author
-
Ryuta Uraki, Maki Kiso, Masaki Imai, Seiya Yamayoshi, Mutsumi Ito, Seiichiro Fujisaki, Emi Takashita, Michiko Ujie, Yuri Furusawa, Atsuhiro Yasuhara, Kiyoko Iwatsuki-Horimoto, Yuko Sakai-Tagawa, Shinji Watanabe, Hideki Hasegawa, and Yoshihiro Kawaoka
- Subjects
Microbiology (medical) ,Mesocricetus ,SARS-CoV-2 ,Immunology ,Antibodies, Monoclonal ,Cell Biology ,Antibodies, Neutralizing ,Antiviral Agents ,Applied Microbiology and Biotechnology ,Microbiology ,COVID-19 Drug Treatment ,Cricetinae ,Genetics ,Animals ,Humans ,RNA, Viral - Abstract
The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the major antigen stimulating the host's protective immune response. Here we assessed the efficacy of therapeutic monoclonal antibodies (mAbs) against Omicron variant (B.1.1.529) sublineage BA.1 variants in Syrian hamsters. Of the FDA-approved therapeutic mAbs tested (that is, REGN10987/REGN10933, COV2-2196/COV2-2130 and S309), only COV2-2196/COV2-2130 efficiently inhibited BA.1 replication in the lungs of hamsters, and this effect was diminished against a BA.1.1 variant possessing the S-R346K substitution. In addition, treatment of BA.1-infected hamsters with molnupiravir (a SARS-CoV-2 RNA-dependent RNA polymerase inhibitor) or S-217622 (a SARS-CoV-2 protease inhibitor) strongly reduced virus replication in the lungs. These findings suggest that the use of therapeutic mAbs in Omicron-infected patients should be carefully considered due to mutations that affect efficacy, and demonstrate that the antiviral compounds molnupiravir and S-217622 are effective against Omicron BA.1 variants.
- Published
- 2022
56. Anti-SARS CoV-2 IgG in COVID-19 Patients with Hematological Diseases: A Single-center, Retrospective Study in Japan
- Author
-
Takayuki, Fujii, Masao, Hagihara, Keiko, Mitamura, Shiori, Nakashima, Shin, Ohara, Tomoyuki, Uchida, Morihiro, Inoue, Moe, Okuda, Atsuhiro, Yasuhara, Jurika, Murakami, Calvin, Duong, Kiyoko, Iwatsuki-Horimoto, Seiya, Yamayoshi, and Yoshihiro, Kawaoka
- Subjects
Adult ,Immunoglobulin M ,Japan ,SARS-CoV-2 ,Immunoglobulin G ,Internal Medicine ,COVID-19 ,Humans ,General Medicine ,Antibodies, Viral ,Hematologic Diseases ,Retrospective Studies - Abstract
Objective Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread globally. Although the relationship between anti-SARS-CoV-2 immunoglobulin G (IgG) antibodies and COVID-19 severity has been reported, information is lacking regarding the seropositivity of patients with particular types of diseases, including hematological diseases. Methods In this single-center, retrospective study, we compared SARS-CoV-2 IgG positivity between patients with hematological diseases and those with non-hematological diseases. Results In total, 77 adult COVID-19 patients were enrolled. Of these, 30 had hematological disorders, and 47 had non-hematological disorders. The IgG antibody against the receptor-binding domain of the spike protein was detected less frequently in patients with hematological diseases (60.0%) than in those with non-hematological diseases (91.5%; p=0.029). Rituximab use was significantly associated with seronegativity (p=0.010). Conclusion Patients with hematological diseases are less likely to develop anti-SARS-CoV-2 antibodies than those with non-hematological diseases, which may explain the poor outcomes of COVID-19 patients in this high-risk group.
- Published
- 2022
57. Efficacy of favipiravir against influenza virus resistant to both baloxavir and neuraminidase inhibitors
- Author
-
Maki Kiso, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Pharmacology ,Microbiology (medical) ,Infectious Diseases ,Pharmacology (medical) - Abstract
Objectives Widespread resistance of influenza viruses to neuraminidase (NA) inhibitor or polymerase inhibitor, baloxavir, is a major public health concern. The amino acid mutations R152K in NA and I38T in polymerase acidic (PA) are responsible for resistance to NA inhibitors and baloxavir, respectively. Methods We generated recombinant A(H1N1)pdm09 viruses possessing NA-R152K, PA-I38T or both mutations by using a plasmid-based reverse genetics system, characterized their virological properties in vitro and in vivo, and examined whether oseltamivir, baloxavir and favipiravir are effective against these mutant viruses. Results The three mutant viruses showed similar or superior growth kinetics and virulence to those of wild-type virus. Although oseltamivir and baloxavir blocked the replication of the wild-type virus in vitro, oseltamivir and baloxavir failed to suppress the replication of the NA-R152K and PA-I38T viruses in vitro, respectively. Mutant virus possessing both mutations grew in the presence of oseltamivir or baloxavir in vitro. Baloxavir treatment protected mice from lethal infection with wild-type or NA-R152K virus, but failed to protect mice from lethal infection with PA-I38T or PA-I38T/NA-R152K virus. Favipiravir treatment protected mice from lethal infection with all viruses tested, whereas oseltamivir treatment did not protect at all. Conclusions Our findings indicate that favipiravir should be used to treat patients with suspected baloxavir-resistant virus infection.
- Published
- 2023
58. Sensitivity of rapid antigen tests for Omicron subvariants of SARS‐CoV‐2
- Author
-
Yuko Sakai‐Tagawa, Seiya Yamayoshi, Peter J. Halfmann, Nancy Wilson, Max Bobholz, William C. Vuyk, Wanting Wei, Hunter Ries, David H. O'Connor, Thomas C. Friedrich, Emilia M. Sordillo, Harm van Bakel, Viviana Simon, and Yoshihiro Kawaoka
- Subjects
Infectious Diseases ,Virology - Published
- 2023
59. The accuracy of reverse genetics systems for SARS‐CoV‐2: Circular polymerase extension reaction versus bacterial artificial chromosome
- Author
-
Yuri Furusawa, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Pulmonary and Respiratory Medicine ,Infectious Diseases ,Epidemiology ,Public Health, Environmental and Occupational Health - Published
- 2023
60. Author response for 'The accuracy of reverse genetics systems for SARS‐CoV‐2: Circular polymerase extension reaction versus bacterial artificial chromosome'
- Author
-
null Yuri Furusawa, null Seiya Yamayoshi, and null Yoshihiro Kawaoka
- Published
- 2023
61. Avian H7N9 influenza viruses are evolutionarily constrained by stochastic processes during replication and transmission in mammals
- Author
-
Katarina M Braun, Luis A Haddock III, Chelsea M Crooks, Gabrielle L Barry, Joseph Lalli, Gabriele Neumann, Tokiko Watanabe, Masaki Imai, Seiya Yamayoshi, Mutsumi Ito, Louise H Moncla, Katia Koelle, Yoshihiro Kawaoka, and Thomas C Friedrich
- Subjects
Virology ,Microbiology - Abstract
H7N9 avian influenza viruses (AIVs) have caused over 1,500 documented human infections since emerging in 2013. Although wild-type H7N9 AIVs can be transmitted by respiratory droplets in ferrets, they have not yet caused widespread outbreaks in humans. Previous studies have revealed molecular determinants of H7N9 AIV host switching, but little is known about potential evolutionary constraints on this process. Here, we compare patterns of sequence evolution for H7N9 AIV and mammalian H1N1 viruses during replication and transmission in ferrets. We show that three main factors—purifying selection, stochasticity, and very narrow transmission bottlenecks—combine to severely constrain the ability of H7N9 AIV to effectively adapt to mammalian hosts in isolated, acute spillover events. We find rare evidence of natural selection favoring new, potentially mammal-adapting mutations within ferrets but no evidence of natural selection acting during transmission. We conclude that human-adapted H7N9 viruses are unlikely to emerge during typical spillover infections. Our findings are instead consistent with a model in which the emergence of a human-transmissible virus would be a rare and unpredictable, though highly consequential, ‘jackpot’ event. Strategies to control the total number of spillover infections will limit opportunities for the virus to win this evolutionary lottery.
- Published
- 2023
62. Efficacy of Antibodies and Antiviral Drugs against Omicron BA.2.12.1, BA.4, and BA.5 Subvariants
- Author
-
Emi Takashita, Seiya Yamayoshi, Viviana Simon, Harm van Bakel, Emilia M. Sordillo, Andrew Pekosz, Shuetsu Fukushi, Tadaki Suzuki, Ken Maeda, Peter Halfmann, Yuko Sakai-Tagawa, Mutsumi Ito, Shinji Watanabe, Masaki Imai, Hideki Hasegawa, and Yoshihiro Kawaoka
- Subjects
SARS-CoV-2 ,Antibodies, Monoclonal ,COVID-19 ,Humans ,Immunologic Factors ,Vaccine Efficacy ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing ,Antiviral Agents ,COVID-19 Drug Treatment - Published
- 2022
63. Efficacy of antivirals and bivalent mRNA vaccines against SARS-CoV-2 isolate CH.1.1
- Author
-
Ryuta Uraki, Mutsumi Ito, Maki Kiso, Seiya Yamayoshi, Kiyoko Iwatsuki-Horimoto, Yuko Sakai-Tagawa, Yuri Furusawa, Masaki Imai, Michiko Koga, Shinya Yamamoto, Eisuke Adachi, Makoto Saito, Takeya Tsutsumi, Amato Otani, Yukie Kashima, Tetsuhiro Kikuchi, Hiroshi Yotsuyanagi, Yutaka Suzuki, and Yoshihiro Kawaoka
- Subjects
Infectious Diseases - Published
- 2023
64. Antiviral and bivalent vaccine efficacy against an omicron XBB.1.5 isolate
- Author
-
Ryuta Uraki, Mutsumi Ito, Maki Kiso, Seiya Yamayoshi, Kiyoko Iwatsuki-Horimoto, Yuri Furusawa, Yuko Sakai-Tagawa, Masaki Imai, Michiko Koga, Shinya Yamamoto, Eisuke Adachi, Makoto Saito, Takeya Tsutsumi, Amato Otani, Tetsuhiro Kikuchi, Hiroshi Yotsuyanagi, Peter J Halfmann, Andrew Pekosz, and Yoshihiro Kawaoka
- Subjects
Infectious Diseases - Published
- 2023
65. Efficacy of Antiviral Agents against the Omicron Subvariant BA.2.75
- Author
-
Emi Takashita, Seiya Yamayoshi, Shuetsu Fukushi, Tadaki Suzuki, Ken Maeda, Yuko Sakai-Tagawa, Mutsumi Ito, Ryuta Uraki, Peter Halfmann, Shinji Watanabe, Makoto Takeda, Hideki Hasegawa, Masaki Imai, and Yoshihiro Kawaoka
- Subjects
SARS-CoV-2 ,COVID-19 ,Humans ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing ,Antiviral Agents ,COVID-19 Drug Treatment - Published
- 2022
66. Previous omicron infection may be protective against reinfection with omicron variant BA.5 for at least 5 months
- Author
-
Michiko, Koga, Kiyoko, Iwatsuki-Horimoto, Tetsuhiro, Kikuchi, Seiya, Yamayoshi, Yoshihiro, Kawaoka, and Hiroshi, Yotsuyanagi
- Subjects
Microbiology (medical) ,Infectious Diseases ,General Medicine - Published
- 2023
67. Characterization of SARS-CoV-2 Omicron BA.2.75 clinical isolates
- Author
-
Ryuta Uraki, Shun Iida, Peter J. Halfmann, Seiya Yamayoshi, Yuichiro Hirata, Kiyoko Iwatsuki-Horimoto, Maki Kiso, Mutsumi Ito, Yuri Furusawa, Hiroshi Ueki, Yuko Sakai-Tagawa, Makoto Kuroda, Tadashi Maemura, Taksoo Kim, Sohtaro Mine, Noriko Iwamoto, Rong Li, Yanan Liu, Deanna Larson, Shuetsu Fukushi, Shinji Watanabe, Ken Maeda, Zhongde Wang, Norio Ohmagari, James Theiler, Will Fischer, Bette Korber, Masaki Imai, Tadaki Suzuki, and Yoshihiro Kawaoka
- Subjects
Multidisciplinary ,General Physics and Astronomy ,General Chemistry ,General Biochemistry, Genetics and Molecular Biology - Abstract
The prevalence of the Omicron subvariant BA.2.75 is rapidly increasing in India and Nepal. In addition, BA.2.75 has been detected in at least 34 other countries and is spreading globally. However, the virological features of BA.2.75 are largely unknown. Here, we evaluated the replicative ability and pathogenicity of BA.2.75 clinical isolates in Syrian hamsters. Although we found no substantial differences in weight change among hamsters infected with BA.2, BA.5, or BA.2.75, the replicative ability of BA.2.75 in the lungs was higher than that of BA.2 and BA.5. Of note, BA.2.75 caused focal viral pneumonia in hamsters, characterized by patchy inflammation interspersed in alveolar regions, which was not observed in BA.5-infected hamsters. Moreover, in competition assays, BA.2.75 replicated better than BA.5 in the lungs of hamsters. These results suggest that BA.2.75 can cause more severe respiratory disease than BA.5 and BA.2 and should be closely monitored.
- Published
- 2022
68. Protective efficacy of a reverse genetics-derived inactivated vaccine against equine influenza virus in horses
- Author
-
Minoru Ohta, Yoshinori Kambayashi, Hiroshi Mita, Taisuke Kuroda, Hiroshi Bannai, Koji Tsujimura, Takashi Yamanaka, Marie Garvey, Ann Cullinane, Seiya Yamayoshi, Yoshihiro Kawaoka, and Manabu Nemoto
- Subjects
Serum Amyloid A Protein ,General Veterinary ,General Immunology and Microbiology ,Fever ,Vaccination ,Public Health, Environmental and Occupational Health ,Neuraminidase ,Antibodies, Viral ,Reverse Genetics ,Influenza A Virus, H3N8 Subtype ,Infectious Diseases ,Hemagglutinins ,Vaccines, Inactivated ,Orthomyxoviridae Infections ,Influenza Vaccines ,Influenza A virus ,Formaldehyde ,Molecular Medicine ,Animals ,Horse Diseases ,Horses - Abstract
Updating vaccine strains is essential to control equine influenza. We evaluated the protective efficacy of an inactivated equine influenza vaccine derived from viruses generated by reverse genetics (RG) in horses in an experimental viral challenge study. Wild-type (WT) virus (A/equine/Tipperary/1/2019) and virus generated by RG (consisting of hemagglutinin and neuraminidase genes from A/equine/Tipperary/1/2019 and six other genes from high-growth A/Puerto Rico/8/34) were inactivated by formalin for vaccine use. Twelve 1-year-old naïve horses with no antibodies against equine influenza virus were assigned to three groups (each n = 4): control, WT, and RG. They were vaccinated twice, 4 weeks apart, and were challenged with A/equine/Tipperary/1/2019 2 weeks after the second vaccination. All four horses in the control group and one horse in the WT group had pyrexia for multiple days and respiratory illness, and one horse in the RG group had pyrexia for 2 days without respiratory illness. The mean rectal temperatures and the mean concentrations of serum amyloid A in the WT and RG groups were significantly lower than those in the control group, with no significant differences between them. The WT and RG vaccines significantly reduced viral shedding relative to the control. The protective efficacy of the RG-derived inactivated vaccine against equine influenza virus is comparable to that of the vaccine derived from WT viruses in horses. The RG technique can make it easy to update equine influenza vaccine strains.
- Published
- 2022
69. Age-stratified seroprevalence of antibodies against SARS-CoV-2 in the pre- and post-vaccination era, February 2020–March 2022, Japan
- Author
-
Seiya Yamayoshi, Kiyoko Iwatsuki-Horimoto, Moe Okuda, Michiko Ujie, Atsuhiro Yasuhara, Jurika Murakami, Calvin Duong, Taiki Hamabata, Mutsumi Ito, Shiho Chiba, Ryo Kobayashi, Satoshi Takahashi, Keiko Mitamura, Masao Hagihara, Akimichi Shibata, Yoshifumi Uwamino, Naoki Hasegawa, Toshiaki Ebina, Akihiko Izumi, Hideaki Kato, Hideaki Nakajima, Norio Sugaya, Yuki Seki, Asef Iqbal, Isamu Kamimaki, Masahiko Yamazaki, Yoshihiro Kawaoka, and Yuki Furuse
- Abstract
Japan has reported a small number of COVID-19 cases relative to other countries. Because not all infected people receive diagnostic tests for COVID-19, the reported number of COVID-19 cases must be lower than the actual number of infections. Assessments of the presence of antibodies against the spike protein of SARS-CoV-2 can retrospectively determine the history of natural infection and vaccination. In this study, we assessed SARS-CoV-2 seroprevalence by analyzing over 60,000 samples collected in Japan from February 2020 to March 2022. The results showed that about 5% of the Japanese population had been infected with the virus by January 2021. The seroprevalence increased with the administration of vaccinations to adults; however, among the elderly, it was not as high as the vaccination rate, probably due to poor immune responses to the vaccines and waning immunity. The infection was spread during the epidemic waves caused by the SARS-CoV-2 Delta and Omicron variants among children who were not eligible for vaccination. Nevertheless, their seroprevalence was as low as 10% as of March 2022. Our study underscores the low incidence of SARS-CoV-2 infection in Japan and the effects of vaccination on immunity at the population level.
- Published
- 2022
70. Characterization of SARS-CoV-2 Omicron BA.4 and BA.5 clinical isolates
- Author
-
Yoshihiro Kawaoka, Ryuta Uraki, Peter Halfmann, Shun Iida, Seiya Yamayoshi, Yuri Furusawa, Maki Kiso, Mutsumi Ito, Kiyoko Iwatsuki-Horimoto, Sohtaro Mine, Makoto Kuroda, Tadashi Maemura, Yuko Sakai, Hiroshi Ueki, Rong Li, Yanan Liu, Deanna Larson, Shuetsu Fukushi, Shinji Watanabe, Ken Maeda, Andrew Pekosz, Ahmed Kandeil, Richard Webby, Zhongde Wang, Masaki Imai, and Tadaki Suzuki
- Abstract
The BA.2 sublineage of the SARS-CoV-2 Omicron variant has become dominant in most countries around the world; however, the prevalence of BA.4 and BA.5 is increasing rapidly in several regions. BA.2 is less pathogenic in animal models than previously circulating variants of concern (VOC). Compared with BA.2, however, BA.4 and BA.5 possess additional substitutions in the spike protein, which play a key role in viral infectivity, raising concerns that the infectivity and pathogenicity of BA.4 and BA.5 are higher than those of BA.2. Here, we evaluated the replicative ability and pathogenicity of authentic BA.4 and BA.5 isolates in wild-type Syrian hamsters and human ACE2 (hACE2) transgenic hamsters. In contrast to recent data with a recombinant chimeric virus possessing the spike protein of BA.4/BA.5 in the background of a BA.2 strain, we observed no obvious differences among BA.2, BA.4, and BA.5 isolates in growth ability or pathogenicity in hamsters, and less pathogenicity compared to a previously circulating Delta (B.1.617.2 lineage) isolate. In addition, in vivo competition experiments revealed that BA.5 outcompeted BA.2 in hamsters, whereas BA.4 and BA.2 exhibited similar fitness. These findings suggest that BA.4 and BA.5 have similar pathogenicity to BA.2 in rodents and that BA.5 possesses viral fitness superior to that of BA.2. Our study highlights the importance of using authentic isolates when evaluating virological features.
- Published
- 2022
71. Characterization of SARS-CoV-2 Omicron BA.4 and BA.5 isolates in rodents
- Author
-
Ryuta Uraki, Peter J. Halfmann, Shun Iida, Seiya Yamayoshi, Yuri Furusawa, Maki Kiso, Mutsumi Ito, Kiyoko Iwatsuki-Horimoto, Sohtaro Mine, Makoto Kuroda, Tadashi Maemura, Yuko Sakai-Tagawa, Hiroshi Ueki, Rong Li, Yanan Liu, Deanna Larson, Shuetsu Fukushi, Shinji Watanabe, Ken Maeda, Andrew Pekosz, Ahmed Kandeil, Richard J. Webby, Zhongde Wang, Masaki Imai, Tadaki Suzuki, and Yoshihiro Kawaoka
- Subjects
Mice ,Multidisciplinary ,Mesocricetus ,SARS-CoV-2 ,Cricetinae ,Spike Glycoprotein, Coronavirus ,Animals ,Humans ,COVID-19 ,Rodentia ,Mice, Transgenic - Abstract
The BA.2 sublineage of the SARS-CoV-2 Omicron variant has become dominant in most countries around the world; however, the prevalence of BA.4 and BA.5 is increasing rapidly in several regions. BA.2 is less pathogenic in animal models than previously circulating variants of concern
- Published
- 2022
72. Characterization of the SARS-CoV-2 B.1.621 (Mu) variant
- Author
-
Peter J. Halfmann, Makoto Kuroda, Tammy Armbrust, James Theiler, Ariane Balaram, Gage K. Moreno, Molly A. Accola, Kiyoko Iwatsuki-Horimoto, Riccardo Valdez, Emily Stoneman, Katarina Braun, Seiya Yamayoshi, Elizabeth Somsen, John J. Baczenas, Keiko Mitamura, Masao Hagihara, Eisuke Adachi, Michiko Koga, Matthew McLaughlin, William Rehrauer, Masaki Imai, Shinya Yamamoto, Takeya Tsutsumi, Makoto Saito, Thomas C. Friedrich, Shelby L. O’Connor, David H. O’Connor, Aubree Gordon, Bette Korber, and Yoshihiro Kawaoka
- Subjects
Membrane Glycoproteins ,SARS-CoV-2 ,Vaccination ,Immunization, Passive ,COVID-19 ,General Medicine ,Antibodies, Viral ,Antibodies, Neutralizing ,Article ,Viral Envelope Proteins ,Neutralization Tests ,Spike Glycoprotein, Coronavirus ,Humans ,COVID-19 Serotherapy - Abstract
The SARS-CoV-2 B.1.621 (Mu) variant emerged in January 2021 and was categorized as a variant of interest by the World Health Organization in August 2021. This designation prompted us to study the sensitivity of this variant to antibody neutralization. In a live virus neutralization assay with serum samples from individuals vaccinated with the Pfizer/BioNTech or Moderna mRNA vaccines, we measured neutralization antibody titers against B.1.621, an early isolate (spike 614D), and a variant of concern (B.1.351, Beta variant). We observed reduced neutralizing antibody titers against the B.1.621 variant (3.4- to 7-fold reduction, depending on the serum sample and time after the second vaccination) compared to the early isolate and a similar reduction when compared to B.1.351. Likewise, convalescent serum from hamsters previously infected with an early isolate neutralized B.1.621 to a lower degree. Despite this antibody titer reduction, hamsters could not be efficiently rechallenged with the B.1.621 variant, suggesting that the immune response to the first infection is adequate to provide protection against a subsequent infection with the B.1.621 variant.
- Published
- 2022
73. Avian H7N9 influenza viruses are evolutionarily constrained by stochastic processes during replication and transmission in mammals
- Author
-
Katarina M. Braun, Luis A. Haddock, Chelsea M. Crooks, Gabrielle L. Barry, Joseph Lalli, Gabriele Neumann, Tokiko Watanabe, Masaki Imai, Seiya Yamayoshi, Mutsumi Ito, Yoshihiro Kawaoka, and Thomas C. Friedrich
- Subjects
virus diseases - Abstract
H7N9 avian influenza viruses (AIV) have caused over 1,500 documented human infections since emerging in 2013. Although wild type H7N9 AIV can transmit by respiratory droplets in ferrets, they have not yet caused widespread outbreaks in humans. Previous studies have revealed molecular determinants of H7N9 AIV virus host-switching, but little is known about potential evolutionary constraints on this process. Here we compare patterns of sequence evolution for H7N9 AIV and mammalian H1N1 viruses during replication and transmission in ferrets. We show that three main factors – purifying selection, stochasticity, and very narrow transmission bottlenecks – combine to severely constrain the ability of H7N9 AIV to effectively adapt to mammalian hosts in isolated, acute spillover events. We find rare evidence of natural selection favoring new or mammalian-adapting mutations within ferrets, but no evidence of natural selection acting during transmission. We conclude that human-adapted H7N9 viruses are unlikely to emerge during typical spillover infections. Our findings are instead consistent with a model in which the emergence of a human-transmissible virus would be a rare and unpredictable, though highly consequential, “jackpot” event. Strategies to limit the total number of spillover infections will limit opportunities for the virus to win this evolutionary lottery.
- Published
- 2022
74. Efficacy of Antiviral Agents against the SARS-CoV-2 Omicron Subvariant BA.2
- Author
-
Emi Takashita, Noriko Kinoshita, Seiya Yamayoshi, Yuko Sakai-Tagawa, Seiichiro Fujisaki, Mutsumi Ito, Kiyoko Iwatsuki-Horimoto, Peter Halfmann, Shinji Watanabe, Kenji Maeda, Masaki Imai, Hiroaki Mitsuya, Norio Ohmagari, Makoto Takeda, Hideki Hasegawa, and Yoshihiro Kawaoka
- Subjects
SARS-CoV-2 ,Spike Glycoprotein, Coronavirus ,Humans ,General Medicine ,Antiviral Agents ,COVID-19 Drug Treatment - Published
- 2022
75. Pathogenesis of Influenza A(H7N9) Virus in Aged Nonhuman Primates
- Author
-
Hiroaki Katsura, Yuriko Tomita, Maki Kiso, Noriko Nakajima, Yoshihiro Kawaoka, Tokiko Watanabe, Jason E. Shoemaker, Hideki Hasegawa, Seiya Yamayoshi, Satoshi Fukuyama, Tadashi Maemura, Robert W. Gregg, Kiyoko Iwatsuki-Horimoto, and Tiago J. S. Lopes
- Subjects
0301 basic medicine ,Aging ,Chemokine ,medicine.medical_treatment ,030106 microbiology ,Inflammation ,Influenza A Virus, H7N9 Subtype ,Virus Replication ,medicine.disease_cause ,Virus ,Proinflammatory cytokine ,Pathogenesis ,Major Articles and Brief Reports ,03 medical and health sciences ,Immune system ,Orthomyxoviridae Infections ,medicine ,Animals ,Immunology and Allergy ,Lung ,biology ,business.industry ,Influenza A virus subtype H5N1 ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Infectious Diseases ,Cytokine ,Immunology ,biology.protein ,Cytokines ,Female ,medicine.symptom ,business - Abstract
The avian influenza A(H7N9) virus has caused high mortality rates in humans, especially in the elderly; however, little is known about the mechanistic basis for this. In the current study, we used nonhuman primates to evaluate the effect of aging on the pathogenicity of A(H7N9) virus. We observed that A(H7N9) virus infection of aged animals (defined as age 20–26 years) caused more severe symptoms than infection of young animals (defined as age 2–3 years). In aged animals, lung inflammation was weak and virus infection was sustained. Although cytokine and chemokine expression in the lungs of most aged animals was lower than that in the lungs of young animals, 1 aged animal showed severe symptoms and dysregulated proinflammatory cytokine and chemokine production. These results suggest that attenuated or dysregulated immune responses in aged animals are responsible for the severe symptoms observed among elderly patients infected with A(H7N9) virus.
- Published
- 2020
76. Characterization and antiviral susceptibility of SARS-CoV-2 Omicron BA.2
- Author
-
Ryuta, Uraki, Maki, Kiso, Shun, Iida, Masaki, Imai, Emi, Takashita, Makoto, Kuroda, Peter J, Halfmann, Samantha, Loeber, Tadashi, Maemura, Seiya, Yamayoshi, Seiichiro, Fujisaki, Zhongde, Wang, Mutsumi, Ito, Michiko, Ujie, Kiyoko, Iwatsuki-Horimoto, Yuri, Furusawa, Ryan, Wright, Zhenlu, Chong, Seiya, Ozono, Atsuhiro, Yasuhara, Hiroshi, Ueki, Yuko, Sakai-Tagawa, Rong, Li, Yanan, Liu, Deanna, Larson, Michiko, Koga, Takeya, Tsutsumi, Eisuke, Adachi, Makoto, Saito, Shinya, Yamamoto, Masao, Hagihara, Keiko, Mitamura, Tetsuro, Sato, Masayuki, Hojo, Shin-Ichiro, Hattori, Kenji, Maeda, Riccardo, Valdez, Moe, Okuda, Jurika, Murakami, Calvin, Duong, Sucheta, Godbole, Daniel C, Douek, Ken, Maeda, Shinji, Watanabe, Aubree, Gordon, Norio, Ohmagari, Hiroshi, Yotsuyanagi, Michael S, Diamond, Hideki, Hasegawa, Hiroaki, Mitsuya, Tadaki, Suzuki, and Lauren, Warsinske
- Subjects
Indazoles ,Lactams ,Proline ,SARS-CoV-2 ,Triazines ,Antibodies, Monoclonal ,COVID-19 ,Cytidine ,Triazoles ,Antibodies, Monoclonal, Humanized ,Antibodies, Viral ,Hydroxylamines ,Antibodies, Neutralizing ,Antiviral Agents ,COVID-19 Drug Treatment ,Drug Combinations ,Mice ,Leucine ,Cricetinae ,Nitriles ,Spike Glycoprotein, Coronavirus ,Animals - Abstract
The recent emergence of SARS-CoV-2 Omicron (B.1.1.529 lineage) variants possessing numerous mutations has raised concerns of decreased effectiveness of current vaccines, therapeutic monoclonal antibodies and antiviral drugs for COVID-19 against these variants
- Published
- 2022
77. Efficacy of Antibodies and Antiviral Drugs against Covid-19 Omicron Variant
- Author
-
Emi Takashita, Noriko Kinoshita, Seiya Yamayoshi, Yuko Sakai-Tagawa, Seiichiro Fujisaki, Mutsumi Ito, Kiyoko Iwatsuki-Horimoto, Shiho Chiba, Peter Halfmann, Hiroyuki Nagai, Makoto Saito, Eisuke Adachi, David Sullivan, Andrew Pekosz, Shinji Watanabe, Kenji Maeda, Masaki Imai, Hiroshi Yotsuyanagi, Hiroaki Mitsuya, Norio Ohmagari, Makoto Takeda, Hideki Hasegawa, and Yoshihiro Kawaoka
- Subjects
Neutralization Tests ,SARS-CoV-2 ,Mutation ,Antibodies, Monoclonal ,Enzyme-Linked Immunosorbent Assay ,General Medicine ,Antiviral Agents ,COVID-19 Drug Treatment - Published
- 2022
78. Therapeutic efficacy of antibodies and antivirals against a SARS-CoV-2 Omicron variant
- Author
-
Ryuta Uraki, Maki Kiso, Masaki Imai, Seiya Yamayoshi, Mutsumi Ito, Michiko Ujie, Yuri Furusawa, Kiyoko Iwatsuki-Horimoto, Yuko Sakai-Tagawa, and Yoshihiro Kawaoka
- Abstract
The spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the major antigen stimulating the host's protective immune response. A new SARS-CoV-2 variant, designated Omicron, first identified in South Africa and possess many spike protein mutations. Here, we assessed the efficacy of therapeutic monoclonal antibodies (mAbs) against an Omicron variant in Syrian hamsters. Of the mAbs tested (i.e., REGN10987/REGN10933, COV2-2196/COV2-2130, and S309), only COV2-2196/COV2-2130 efficiently inhibited the replication of the Omicron variant in the lungs of hamsters. We also found that treatment of Omicron-infected hamsters with molnupiravir (an inhibitor of the RNA-dependent RNA polymerase of SARS-CoV-2) or S-217622 (an inhibitor of the main protease of SARS-CoV-2) led to a dramatic reduction of virus replication in the lungs. These findings suggest that treatment with the mAb combination COV2-2196/COV2-2130 or the antiviral compounds molnupiravir and S-217622 may be effective against the Omicron variants.
- Published
- 2022
79. The SARS-CoV-2 B.1.1.529 Omicron virus causes attenuated infection and disease in mice and hamsters
- Author
-
Michael Diamond, Peter Halfmann, Tadashi Maemura, Kiyoko Iwatsuki-Horimoto, Shun Iida, Maki Kiso, Suzanne Scheaffer, Tamarand Darling, Astha Joshi, Samantha Loeber, Stephanie Foster, Baoling Ying, Bradley Whitener, Katharine Floyd, Michiko Ujie, Noriko Nakajima, Mutsumi Ito, Ryan Wright, Ryuta Uraki, Rong Li, Yuko Sakai, Yanan Liu, Deanna Larson, Jorge Osorio, Juan Hernandez-Ortiz, Karl Čiuoderis, Kelsey Florek, Mit Patel, Allen Bateman, Abby Odle, Lok-Yin Wong, Zhongde Wang, Venkata Viswanadh Edara, Zhenlu Chong, Larissa Thackray, Hiroshi Ueki, Seiya Yamayoshi, Masaki Imai, Stanley Perlman, Richard Webby, Robert Seder, Mehul Suthar, Adolfo Garcia-Sastre, Michael Schotsaert, Tadaki Suzuki, Adrianus Boon, Yoshihiro Kawaoka, Daniel Douek, Juan Moliva, Nancy Sullivan, Matthew Gagne, Amy Ransier, James Case, Trushar Jeevan, John Franks, Thomas Fabrizio, Jennifer DeBeauchamp, Lisa Kercher, Patrick Seiler, Gagandeep Singh, Prajakta Warang, Ana S. Gonzalez-Reiche, Emilia Sordillo, Harm van Bakel, and Viviana Simon
- Abstract
Despite the development and deployment of antibody and vaccine countermeasures, rapidly-spreading SARS-CoV-2 variants with mutations at key antigenic sites in the spike protein jeopardize their efficacy. The recent emergence of B.1.1.529, the Omicron variant1,2, which has more than 30 mutations in the spike protein, has raised concerns for escape from protection by vaccines and therapeutic antibodies. A key test for potential countermeasures against B.1.1.529 is their activity in pre-clinical rodent models of respiratory tract disease. Here, using the collaborative network of the SARS-CoV-2 Assessment of Viral Evolution (SAVE) program of the National Institute of Allergy and Infectious Diseases (NIAID), we evaluated the ability of multiple B.1.1.529 Omicron isolates to cause infection and disease in immunocompetent and human ACE2 (hACE2) expressing mice and hamsters. Despite modeling and binding data suggesting that B.1.1.529 spike can bind more avidly to murine ACE2, we observed attenuation of infection in 129, C57BL/6, and BALB/c mice as compared with previous SARS-CoV-2 variants, with limited weight loss and lower viral burden in the upper and lower respiratory tracts. Although K18-hACE2 transgenic mice sustained infection in the lungs, these animals did not lose weight. In wild-type and hACE2 transgenic hamsters, lung infection, clinical disease, and pathology with B.1.1.529 also were milder compared to historical isolates or other SARS-CoV-2 variants of concern. Overall, experiments from multiple independent laboratories of the SAVE/NIAID network with several different B.1.1.529 isolates demonstrate attenuated lung disease in rodents, which parallels preliminary human clinical data.
- Published
- 2022
80. Baloxavir Marboxil Treatment of Nude Mice Infected With Influenza A Virus
- Author
-
Jurika Murakami, Yoshihiro Kawaoka, Maki Kiso, and Seiya Yamayoshi
- Subjects
Dibenzothiepins ,Male ,Pyridones ,Morpholines ,Orthomyxoviridae ,Mutant ,Mice, Nude ,Drug resistance ,medicine.disease_cause ,Antiviral Agents ,Virus ,Mice ,Major Articles and Brief Reports ,Orthomyxoviridae Infections ,Influenza A virus ,medicine ,Animals ,Immunology and Allergy ,Respiratory system ,Dose-Response Relationship, Drug ,biology ,Triazines ,business.industry ,biology.organism_classification ,Virology ,Infectious Diseases ,medicine.anatomical_structure ,biology.protein ,business ,Neuraminidase ,Respiratory tract - Abstract
Background Immunocompromised patients infected with influenza virus require prolonged treatment with neuraminidase inhibitors, because these patients are not able to eradicate the virus from the respiratory tract, leading to the emergence of drug-resistant mutant viruses. Methods In this study, we examined the efficacy of baloxavir marboxil in nude mice that were immunologically deficient. Results Daily treatment with a suboptimal dose of baloxavir marboxil increased the survival time of the virus-infected nude mice but did not clear the virus from their respiratory organs, resulting in gradual body weight loss after termination of treatment. Conclusions Despite the prolonged baloxavir marboxil treatment, few resistant mutants were detected.
- Published
- 2019
81. Immunological Correlates of Prevention of the Onset of Seasonal H3N2 Influenza
- Author
-
Moe Okuda, Yuko Sakai-Tagawa, Michiko Koga, Tomohiko Koibuchi, Tadashi Kikuchi, Eisuke Adachi, Lay Ahyoung Lim, Shinya Yamamoto, Hiroshi Yotsuyanagi, Kyota Negishi, Daisuke Jubishi, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Infectious Diseases ,Influenza A Virus, H1N1 Subtype ,Hemagglutinins ,Influenza Vaccines ,Influenza A Virus, H3N2 Subtype ,Influenza, Human ,Major Article ,Immunology and Allergy ,Humans ,Neuraminidase ,Hemagglutinin Glycoproteins, Influenza Virus ,Seasons ,Antibodies, Viral - Abstract
On influenza virus infection or vaccination, immune responses occur, including the production of antibodies with various functions that contribute to protection from seasonal influenza virus infection. In the current study, we attempted to identify the antibody functions that play a central role in preventing the onset of seasonal influenza by comparing the levels of several antibody titers for different antibody functions between 5 subclinically infected individuals and 16 patients infected with seasonal H3N2 virus. For antibody titers before influenza virus exposure, we found that the nAb titers and enzyme-linked immunosorbent assay titers against hemagglutinin and neuraminidase (NA) proteins in the subclinically infected individuals were significantly higher than those in the patients, whereas the NA inhibition titers and antibody-dependent cellular cytotoxicity activities did not significantly differ between subclinically infected individuals and infected patients. These results suggest that nAb and enzyme-linked immunosorbent assay titers against hemagglutinin and NA serve as correlates of symptomatic influenza infection.
- Published
- 2021
82. Non-propagative human parainfluenza virus type 2 nasal vaccine robustly protects the upper and lower airways against SARS-CoV-2
- Author
-
Mitsuyo Maeda, Yoshihiro Kawaoka, Mutsumi Ito, Ryoichi Ono, Masaki Imai, Seiya Yamayoshi, Junpei Ohtsuka, Yosky Kataoka, Tadashi Maemura, Tetsuya Nosaka, Asami Eguchi, and Masayuki Fukumura
- Subjects
Multidisciplinary ,viruses ,Science ,RNA ,Infection control in health technology ,Biology ,Virology ,Immunoglobulin G ,Article ,Vaccination ,Viral envelope ,biology.protein ,Nasal administration ,Vector (molecular biology) ,Antibody ,Gene - Abstract
We developed an intranasal vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using the replication-incompetent human parainfluenza virus type 2 (hPIV2) vector BC-PIV, which can deliver ectopic gene as stable RNA and ectopic protein on the envelope. BC-PIV expressing the full-length prefusion-stabilized spike gene (K986P/V987P) of SARS-CoV-2, S-2PM, possessed a corona-like viral envelope. Intranasal vaccination of mice with BC-PIV/S-2PM induced high levels of neutralizing immunoglobulin G (IgG) and mucosal IgA antibodies against the spike protein. Although BC-PIV showed hemagglutinating activity, BC-PIV/S-2PM lacked such activity, in accordance with the presence of the massive spike protein on the viral surface. Furthermore, single-dose intranasal vaccination of hamsters with BC-PIV/S-2PM completely protected the lungs from SARS-CoV-2 at 11-week post-immunization, and boost vaccination two weeks before the challenge conferred virtually complete protection of the nasal turbinates against SARS-CoV-2. Thus, this chimeric hPIV2/spike intranasal vaccine is a promising vaccine candidate for SARS-CoV-2 to curtail virus transmission., Graphical abstract, Infection control in health technology; Virology
- Published
- 2021
83. Antibody-Dependent Enhancement of SARS-CoV-2 Infection Is Mediated by the IgG Receptors FcγRIIA and FcγRIIIA but Does Not Contribute to Aberrant Cytokine Production by Macrophages
- Author
-
Seiya Yamayoshi, Makoto Kuroda, Peter Halfmann, Tammy Armbrust, Yoshihiro Kawaoka, and Tadashi Maemura
- Subjects
Chemokine ,Feline coronavirus ,viruses ,medicine.medical_treatment ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Proinflammatory cytokine ,Immune system ,Cricetinae ,Virology ,medicine ,Animals ,Humans ,Antibody-dependent enhancement ,FcγRIIA ,biology ,SARS-CoV-2 ,Macrophages ,Receptors, IgG ,COVID-19 ,Antibody-Dependent Enhancement ,QR1-502 ,FcγRIIIA ,Cytokine ,Immunology ,biology.protein ,Cytokines ,Antibody ,ADE ,Research Article - Abstract
The coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the detrimental effects of antibodies. Antibody-dependent enhancement (ADE) of infection is one of the biggest concerns in terms of not only the antibody reaction to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) upon reinfection with the virus but also the reaction to COVID-19 vaccines. In this study, we evaluated ADE of infection by using COVID-19 convalescent-phase plasma and BHK cells expressing human Fcγ receptors (FcγRs). We found that FcγRIIA and FcγRIIIA mediated modest ADE of infection against SARS-CoV-2. Although ADE of infection was observed in monocyte-derived macrophages infected with SARS-CoV-2, including its variants, proinflammatory cytokine/chemokine expression was not upregulated in macrophages. SARS-CoV-2 infection thus produces antibodies that elicit ADE of infection, but these antibodies do not contribute to excess cytokine production by macrophages. IMPORTANCE Viruses infect cells mainly via specific receptors at the cell surface. Antibody-dependent enhancement (ADE) of infection is an alternative mechanism of infection for viruses to infect immune cells that is mediated by antibodies and IgG receptors (FcγRs). Because ADE of infection contributes to the pathogenesis of some viruses, such as dengue virus and feline coronavirus, it is important to evaluate the precise mechanism of ADE and its contribution to the pathogenesis of SARS-CoV-2. Here, using convalescent-phase plasma from COVID-19 patients, we found that two types of FcγRs, FcγRIIA and FcγRIIIA, mediate ADE of SARS-CoV-2 infection. Although ADE of infection was observed for SARS-CoV-2 and its recent variants, proinflammatory cytokine production in monocyte-derived macrophages was not upregulated. These observations suggest that SARS-CoV-2 infection produces antibodies that elicit ADE of infection, but these antibodies may not be involved in aberrant cytokine release by macrophages during SARS-CoV-2 infection.
- Published
- 2021
84. Comparative Sensitivity of Rapid Antigen Tests for the Delta Variant (B.1.617.2) of SARS-CoV-2
- Author
-
Yoshihiro Kawaoka, Yuko Sakai-Tagawa, Peter Halfmann, and Seiya Yamayoshi
- Subjects
Delta ,Saliva ,delta variant ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Biology ,Microbiology ,Sensitivity and Specificity ,Virus ,Article ,COVID-19 Serological Testing ,Antigen ,Virology ,Humans ,rapid antigen test ,Antigens, Viral ,Infectious virus ,Reagent Strips ,Immunoassay ,B.1.617.2 ,SARS-CoV-2 ,COVID-19 ,sensitivity ,QR1-502 ,Infectious Diseases ,Rapid antigen test - Abstract
Rapid antigen tests (RATs) for COVID-19 based on lateral flow immunoassays are useful for rapid diagnosis in a variety of settings. Although many kinds of RATs are available, their respective sensitivity has not been compared. Here, we examined the sensitivity of 27 RATs available in Japan for the detection of the SARS-CoV-2 delta variant. All of the RATs tested detected the delta variant albeit with different sensitivities. Nine RATs (ESPLINE SARS-CoV-2, ALSONIC COVID-19 Ag, COVID-19 and Influenza A+B Antigen Combo Rapid Test, ImmunoArrow SARS-CoV-2, Fuji Dri-chem immuno AG cartridge COVID-19 Ag, 2019-nCoV Ag rapid detection kit, Saliva SARS-CoV-2(2019-nCoV) Antigen Test Kit, and Rabliss SARS-CoV-2 antigen detection kit COVID19 AG) showed superior sensitivity to the isolated delta variant. Although actual clinical specimens were not examined, the detection level of most of the RATs was 7500 pfu, indicating that individuals whose test samples contained less virus than that would be considered negative. Therefore, it is important to bear in mind that RATs may miss individuals shedding low levels of infectious virus.
- Published
- 2021
85. A method to reduce ELISA serial dilution assay workload applied to SARS-CoV-2 and seasonal HCoVs
- Author
-
Robert Presler, Peter J. Jester, Gabriele Neumann, Hongyu Rao, Kiyoko Iwatsuki-Horimoto, Yoshihiro Kawaoka, Masao Hagihara, Keiko Mitamura, Shiho Chiba, Tomoyuki Uchida, Seiya Yamayoshi, Lizheng Guan, Nobuhiro Ikeda, Peter Halfmann, and David J. Pattinson
- Subjects
Titer ,Antigen ,Serial dilution ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,biology.protein ,Spike Protein ,Biology ,Antibody ,Serum samples ,Virology - Abstract
ObjectivesAssays using ELISA measurements on serially diluted serum samples have been heavily used to measure serum reactivity to SARS-CoV-2 antigens and are widely used in virology and elsewhere in biology. We test a method to reduce the workload of these assays, and measure reactivity of SARS-CoV-2 and HCoV antigens to human serum samples collected before and during the COVID-19 pandemic.MethodsWe apply Bayesian hierarchical modelling to ELISA measurements of human serum samples against SARS-CoV-2 and HCoV antigens.ResultsInflection titers for SARS-CoV-2 full-length spike protein (S1S2), spike protein receptor-binding domain (RBD), and nucleoprotein (N) inferred from three spread-out dilutions correlated with those inferred from eight consecutive dilutions with an R2 value of 0.97 or higher. We confirm existing findings showing a small proportion of pre-pandemic human serum samples contain cross-reactive antibodies to SARS-CoV-2 S1S2 and N, and that SARS-CoV-2 infection increases serum reactivity to the beta-HCoVs OC43 and HKU1 S1S2.ConclusionsIn serial dilution assays, large savings in resources and/or increases in throughput can be achieved by reducing the number of dilutions measured and using Bayesian hierarchical modelling to infer inflection or endpoint titers. We have released software for conducting these types of analysis.
- Published
- 2021
86. Characterization of a new SARS-CoV-2 variant that emerged in Brazil
- Author
-
Hiroaki Mitsuya, Tadashi Maemura, Ichiro Nakachi, Rie Baba, Riccardo Valdez, Hiroyuki Nagai, Hideaki Nakajima, Tetsuya Suzuki, Michiko Koga, Junichi Ochi, Tadaki Suzuki, Aubree Gordon, Samantha Loeber, Kensuke Fujita, Kenji Maeda, Maki Kiso, Carmen Gherasim, David A. Baker, Norio Sugaya, Makoto Saito, Noriko Nakajima, Yoshihiro Kawaoka, Takayuki Ogura, John J. Baczenas, Moe Okuda, Hideaki Kato, Kiyoko Iwatsuki-Horimoto, Shuetsu Fukushi, Hiroshi Ueki, Tiago J. S. Lopes, Yudai Kuroda, Shiho Chiba, Tokiko Watanabe, Makoto Kuroda, Norio Ohmagari, Tsuguto Fujimoto, Masaki Imai, Osamu Akasaka, Peter Halfmann, Masato Hatta, Michiko Ujie, Eisuke Adachi, Ken Maeda, Shelby L. O’Connor, Hiroshi Yotsuyanagi, Atsuhiro Yasuhara, Shinya Yamamoto, Amie J. Eisfeld, Yusuke Miyazato, Kenta Takahashi, Keiko Mitamura, Yuko Sakai-Tagawa, Mutsumi Ito, Shin ichiro Hattori, Morio Nakamura, Yuri Furusawa, David H. O’Connor, Seiya Yamayoshi, and Kazuma Yagi
- Subjects
0106 biological sciences ,0301 basic medicine ,Letter ,viruses ,Virus Replication ,Microbiology ,01 natural sciences ,Neutralization ,reinfection ,Mice ,03 medical and health sciences ,Immunogenicity, Vaccine ,Immune system ,Antigen ,Cricetinae ,Animals ,Humans ,skin and connective tissue diseases ,Lung ,Infectivity ,Multidisciplinary ,Mesocricetus ,biology ,SARS-CoV-2 ,fungi ,virus diseases ,COVID-19 ,convalescent human plasma ,X-Ray Microtomography ,Biological Sciences ,biology.organism_classification ,Antibodies, Neutralizing ,Virology ,body regions ,Titer ,030104 developmental biology ,P.1 variant ,Viral replication ,Spike Glycoprotein, Coronavirus ,biology.protein ,Antibody ,Syrian hamsters ,010606 plant biology & botany - Abstract
Significance Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants are of concern, with the P.1 variants dominating in Brazil. Brazil is now seeing a record number of deaths. Here, we report that the pathogenicity in hamsters of a P.1 variant is similar to that of nonvariant SARS-CoV-2. However, it has an expanded host range as shown by its replication in mice. Prior infection with nonvariant SARS-CoV-2 strains efficiently prevented replication of the P.1 variant in the lower respiratory tract of hamsters upon reinfection. Convalescent sera from patients infected with nonvariants or sera from messenger RNA vaccinees showed comparable neutralization titers among the P.1 and previously circulating strains. These results suggest that previous SARS-CoV-2 infection and vaccines based on the original SARS-CoV-2 will provide some protection against P.1 infection., The spike (S) protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) plays a key role in viral infectivity. It is also the major antigen stimulating the host's protective immune response, specifically, the production of neutralizing antibodies. Recently, a new variant of SARS-CoV-2 possessing multiple mutations in the S protein, designated P.1, emerged in Brazil. Here, we characterized a P.1 variant isolated in Japan by using Syrian hamsters, a well-established small animal model for the study of SARS-CoV-2 disease (COVID-19). In hamsters, the variant showed replicative abilities and pathogenicity similar to those of early and contemporary strains (i.e., SARS-CoV-2 bearing aspartic acid [D] or glycine [G] at position 614 of the S protein). Sera and/or plasma from convalescent patients and BNT162b2 messenger RNA vaccinees showed comparable neutralization titers across the P.1 variant, S-614D, and S-614G strains. In contrast, the S-614D and S-614G strains were less well recognized than the P.1 variant by serum from a P.1-infected patient. Prior infection with S-614D or S-614G strains efficiently prevented the replication of the P.1 variant in the lower respiratory tract of hamsters upon reinfection. In addition, passive transfer of neutralizing antibodies to hamsters infected with the P.1 variant or the S-614G strain led to reduced virus replication in the lower respiratory tract. However, the effect was less pronounced against the P.1 variant than the S-614G strain. These findings suggest that the P.1 variant may be somewhat antigenically different from the early and contemporary strains of SARS-CoV-2.
- Published
- 2021
87. SARS-CoV-2 spike P681R mutation enhances and accelerates viral fusion
- Author
-
Masaki Imai, Isao Yoshida, Takashi Irie, Kiyoko Iwatsuki-Horimoto, Seiya Ozono, Ryoko Kawabata, Takasuke Fukuhara, So Nakagawa, Seiya Yamayoshi, Mami Nagashima, Yoshihito Horisawa, Yuri L Tanaka, Terumasa Ikeda, Rigel Suzuki, Erika P Butlertanaka, Akifumi Takaori-Kondo, Ryosuke Nomura, Izumi Kimura, Takemasa Sakaguchi, Jiaqi Wu, Hesham Nasser, Mutsumi Ito, Ryo Shimizu, Jumpei Ito, Keiya Uriu, Kotaro Shirakawa, Yusuke Kosugi, Kazuhisa Yoshimura, Yasuhiro Kazuma, Kenzo Tokunaga, Akatsuki Saito, Kumiko Yoshimatsu, Kenji Sadamasu, Kenta Shimizu, Yoshihiro Kawaoka, Tadashi Maemura, Kei Sato, and Hiroyuki Asakura
- Subjects
Delta ,Mutation ,Lineage (genetic) ,medicine ,Spike (database) ,Biology ,Pathogenicity ,medicine.disease_cause ,Virology ,Genome ,Phenotype ,Virus - Abstract
SummaryDuring the current SARS-CoV-2 pandemic, a variety of mutations have been accumulated in the viral genome, and at least five variants of concerns (VOCs) have been considered as the hazardous SARS-CoV-2 variants to the human society. The newly emerging VOC, the B.1.617.2 lineage (delta variant), closely associates with a huge COVID-19 surge in India in Spring 2021. However, its virological property remains unclear. Here, we show that the B.1.617 variants are highly fusogenic and form prominent syncytia. Bioinformatic analyses reveal that the P681R mutation in the spike protein is highly conserved in this lineage. Although the P681R mutation decreases viral infectivity, this mutation confers the neutralizing antibody resistance. Notably, we demonstrate that the P681R mutation facilitates the furin-mediated spike cleavage and enhances and accelerates cell-cell fusion. Our data suggest that the P681R mutation is a hallmark characterizing the virological phenotype of this newest VOC, which may associate with viral pathogenicity.HighlightsP681R mutation is highly conserved in the B.1.617 lineagesP681R mutation accelerates and enhances SARS-CoV-2 S-mediated fusionPromotion of viral fusion by P681R mutation is augmented by TMPRSS2
- Published
- 2021
88. Uncovering the Anti-Ebola Repertome
- Author
-
Seiya Yamayoshi and Yoshihiro Kawaoka
- Subjects
Disease onset ,Proteome ,Disease ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,VP40 ,Antigen ,Virology ,medicine ,Humans ,Survivors ,030304 developmental biology ,Ebolavirus ,0303 health sciences ,biology ,Hemorrhagic Fever, Ebola ,humanities ,Antibody response ,biology.protein ,Parasitology ,Antibody ,030217 neurology & neurosurgery - Abstract
Evolution of antibody repertoire against the Ebola virus (EBOV) proteome was characterized in an acutely infected patient receiving supportive care alone to elucidate virus-host interactions over time. Differential kinetics are observed for IgM/IgG/IgA epitope diversity, antibody binding, and affinity maturation to EBOV proteins. During acute illness, antibodies predominate to VP40 and Glycoprotein (GP). At day 13 of clinical illness a marked increase in antibody titers to most EBOV proteins and affinity maturation to GP is associated with rapid decline in viral replication and illness severity. At one-year, despite undetectable virus, a diverse IgM repertoire against VP40 and GP epitopes is observed suggesting occult viral persistence. Rabbit immunization experiments identify key immunodominant sites of GP, while challenge studies in mice found these epitopes induce EBOV-neutralizing antibodies and protect against lethal EBOV challenge. This study reveals markers of viral persistence and provides promising approaches for development and evaluation of vaccines and therapeutics.
- Published
- 2020
89. Characterization of H7N9 avian influenza viruses isolated from duck meat products
- Author
-
Huapeng Feng, Maki Kiso, Yoshihiro Kawaoka, Akihiro Shibata, Hiromichi Mitake, Kiyoko Iwatsuki‐Hirimoto, Mutsumi Ito, Seiya Yamayoshi, Tiago J. S. Lopes, Hiroyuki Osaka, Tokiko Watanabe, Li Wu, Riho Sumiyoshi, and Masaki Imai
- Subjects
040301 veterinary sciences ,animal diseases ,viruses ,Highly pathogenic ,Chick Embryo ,Biology ,Influenza A Virus, H7N9 Subtype ,medicine.disease_cause ,Virus ,Madin Darby Canine Kidney Cells ,0403 veterinary science ,Mice ,03 medical and health sciences ,Dogs ,Japan ,Influenza, Human ,medicine ,Animals ,Humans ,Poultry Products ,Poultry Diseases ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,General Veterinary ,General Immunology and Microbiology ,Ferrets ,virus diseases ,04 agricultural and veterinary sciences ,General Medicine ,Pathogenicity ,Virology ,Low pathogenic ,Influenza A virus subtype H5N1 ,Ducks ,Influenza in Birds ,Female - Abstract
Avian influenza H7N9 viruses have caused five epidemic waves of human infections since the first human cases were reported in 2013. In 2016, the initial low pathogenic avian influenza (LPAI) H7N9 viruses became highly pathogenic, acquiring multi-basic amino acids at the haemagglutinin cleavage site. These highly pathogenic avian influenza (HPAI) H7N9 viruses have been detected in poultry and humans in China, causing concerns of a serious threat to global public health. In Japan, both HPAI and LPAI H7N9 viruses were isolated from duck meat products carried illegally and relinquished voluntarily at the border by passengers on flights from China to Japan between 2016 and 2017. Some of the LPAI and HPAI H7N9 viruses detected at the border in Japan were characterized previously in chickens and ducks; however, their pathogenicity and replicative ability in mammals remain unknown. In this study, we assessed the biological features of two HPAI H7N9 virus isolates [A/duck/Japan/AQ-HE29-22/2017 (HE29-22) and A/duck/Japan/AQ-HE29-52/2017 (HE29-52); both of these viruses were isolated from duck meat at the border)] and an LPAI H7N9 virus isolate [A/duck/Japan/AQ-HE28-3/2016 (HE28-3)] in mice and ferrets. In mice, HE29-52 was more pathogenic than HE29-22 and HE28-3. In ferrets, the two HPAI virus isolates replicated more efficiently in the lower respiratory tract of the animals than did the LPAI virus isolate. Our results indicate that HPAI H7N9 viruses with the potential to cause severe diseases in mammals have been illegally introduced to Japan.
- Published
- 2019
90. A single amino acid change in hemagglutinin reduces the cross-reactivity of antiserum against an equine influenza vaccine strain
- Author
-
Seiya Yamayoshi, Hiroshi Bannai, Takashi Yamanaka, Koji Tsujimura, Manabu Nemoto, Yoshihiro Kawaoka, and Hiroshi Kokado
- Subjects
Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Cross Reactions ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Cross-reactivity ,03 medical and health sciences ,Orthomyxoviridae Infections ,Virology ,medicine ,Animals ,Horses ,Pathogen ,030304 developmental biology ,Antiserum ,0303 health sciences ,030306 microbiology ,Immune Sera ,Antibody titer ,General Medicine ,Reverse genetics ,Vaccination ,Amino Acid Substitution ,Influenza A virus ,Influenza Vaccines ,biology.protein ,Horse Diseases ,Antibody - Abstract
Equine influenza virus is an important pathogen for the horse industry because of its economic impact, and vaccination is a key control measure. Our previous work suggested that a mutation at position 144 in the hemagglutinin of Florida sublineage clade 2 viruses reduces the cross-neutralizing activity of antiserum against a former vaccine strain. To confirm this suggestion, here, we generated viruses by reverse genetics. Antibody titers against the mutated viruses were one-tenth to one-sixteenth of those against the former vaccine strain. Our findings confirm that this single amino acid substitution reduces the cross-reactivity of antiserum against this former Japanese vaccine.
- Published
- 2019
91. Antibody-free digital influenza virus counting based on neuraminidase activity
- Author
-
Yoichiro Fujioka, Seiya Yamayoshi, Kazuhito V. Tabata, Yusuke Ohba, Yoshihiro Kawaoka, Yoshiki Moriizumi, Yoshihiro Minagawa, Yuko Kawaguchi, Hiroyuki Noji, and Mana Ono
- Subjects
0301 basic medicine ,Oseltamivir ,viruses ,Neuraminidase ,lcsh:Medicine ,medicine.disease_cause ,Virus ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Proteins ,0302 clinical medicine ,Influenza A Virus, H1N1 Subtype ,Influenza A virus ,medicine ,lcsh:Science ,Detection limit ,Multidisciplinary ,biology ,Chemistry ,Influenza A Virus, H3N2 Subtype ,lcsh:R ,Virion ,Virology ,Titer ,030104 developmental biology ,biology.protein ,Alkaline phosphatase ,lcsh:Q ,Antibody ,030217 neurology & neurosurgery - Abstract
There is large demand for a quantitative method for rapid and ultra-sensitive detection of the influenza virus. Here, we established a digital influenza virus counting (DIViC) method that can detect a single virion without antibody. In the assay, a virion is stochastically entrapped inside a femtoliter reactor array device for the fluorogenic assay of neuraminidase, and incubated for minutes. By analyzing 600,000 reactors, the practical limit of detection reached the order of 103 (PFU)/mL, only 10-times less sensitive than RT-PCR and more than 1000-times sensitive than commercial rapid test kits (RIDTs). Interestingly, neuraminidase activity differed among virions. The coefficient of variance was 30–40%, evidently broader than that of alkaline phosphatase measured as a model enzyme for comparison, suggesting the heterogeneity in size and integrity among influenza virus particles. Sensitivity to oseltamivir also differed between virions. We also tested DIViC using clinical gargle samples that imposes less burden for sampling while with less virus titre. The comparison with RIDTs showed that DIViC was largely superior to RIDTs in the sensitivity with the clinical samples although a few false-positive signals were observed in some clinical samples that remains as a technical challenge.
- Published
- 2019
92. Optimization of an LNP-mRNA vaccine candidate targeting SARS-CoV-2 receptor-binding domain
- Author
-
Yasushi Itoh, Kiyoko Iwatsuki-Horimoto, Mutsumi Ito, Nao Jounai, Hioki Kou, Seiya Yamayoshi, Yoshinori Kitagawa, Takako Niwa, Fumihiko Takeshita, Takashi Suzuki, Yoshihiro Kawaoka, Misako Nakayama, Kouji Kobiyama, Ken Ishii, Shintaro Yamada, Temizoz B, Negishi H, Tsuchida J, Masayuki Imai, Cong Thanh Nguyen, Tomoyuki Watanabe, Maki Kiso, and Hirohito Ishigaki
- Subjects
Messenger RNA ,Reactogenicity ,Innate immune system ,Immunization ,Interferon ,Immunogenicity ,medicine ,Biology ,medicine.disease_cause ,Type I interferon production ,Virology ,Coronavirus ,medicine.drug - Abstract
In 2020, two mRNA-based vaccines, encoding the full length of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, have been introduced for control of the coronavirus disease (COVID-19) pandemic1,2. However, reactogenicity, such as fever, caused by innate immune responses to the vaccine formulation remains to be improved. Here, we optimized a lipid nanoparticle (LNP)-based mRNA vaccine candidate, encoding the SARS-CoV-2 spike protein receptor-binding domain (LNP-mRNA-RBD), which showed improved immunogenicity by removing reactogenic materials from the vaccine formulation and protective potential against SARS-CoV-2 infection in cynomolgus macaques. LNP-mRNA-RBD induced robust antigen-specific B cells and follicular helper T cells in the BALB/c strain but not in the C57BL/6 strain; the two strains have contrasting abilities to induce type I interferon production by dendritic cells. Removal of reactogenic materials from original synthesized mRNA by HPLC reduced type I interferon (IFN) production by dendritic cells, which improved immunogenicity. Immunization of cynomolgus macaques with an LNP encapsulating HPLC-purified mRNA induced robust anti-RBD IgG in the plasma and in various mucosal areas, including airways, thereby conferring protection against SARS-CoV-2 infection. Therefore, fine-tuning the balance between the immunogenic and reactogenic activity of mRNA-based vaccine formulations may offer safer and more efficacious outcomes.
- Published
- 2021
93. Author response for 'Growth properties and immunogenicity of a virus generated by reverse genetics for an inactivated equine influenza vaccine'
- Author
-
Minoru Ohta, Hiroshi Bannai, Yoshihiro Kawaoka, Norihisa Tamura, Yoshinori Kambayashi, Koji Tsujimura, Seiya Yamayoshi, and Manabu Nemoto
- Subjects
Immunogenicity ,Equine influenza vaccine ,Biology ,Virology ,Reverse genetics ,Virus - Published
- 2021
94. Growth properties and immunogenicity of a virus generated by reverse genetics for an inactivated equine influenza vaccine
- Author
-
Seiya Yamayoshi, Norihisa Tamura, Manabu Nemoto, Yoshinori Kambayashi, Minoru Ohta, Hiroshi Bannai, Koji Tsujimura, and Yoshihiro Kawaoka
- Subjects
040301 veterinary sciences ,Equine influenza ,Antibodies, Viral ,Virus ,0403 veterinary science ,Orthomyxoviridae Infections ,Animals ,Horses ,biology ,Inoculation ,Immunogenicity ,Vaccination ,0402 animal and dairy science ,Embryonated ,04 agricultural and veterinary sciences ,General Medicine ,040201 dairy & animal science ,Virology ,Reverse Genetics ,Influenza Vaccines ,Inactivated vaccine ,Viruses ,biology.protein ,Horse Diseases ,Neuraminidase - Abstract
Background Keeping vaccine strains up to date is the key to controlling equine influenza (EI). Viruses generated by reverse genetics (RG) are likely to be effective for quickly updating a vaccine strain. Objectives To evaluate the growth properties of an RG virus in embryonated chicken eggs, and to evaluate antibody responses to a formalin-inactivated vaccine derived from the RG virus in Thoroughbred horses. Study design In vitro and in vivo experiments. Methods Wild-type (WT) viruses (A/equine/Ibaraki/1/2007) or RG viruses (consisting of haemagglutinin [HA] and neuraminidase genes derived from A/equine/Ibaraki/1/2007 and the six other genes derived from high-growth A/Puerto Rico/8/34) were inoculated into embryonated chicken eggs, and the allantoic fluids were harvested at every 24 hours after inoculation. WT and RG viruses were inactivated by formalin for vaccine use. Ten unvaccinated yearlings (five each for WT or RG vaccine) received the first two doses of a primary vaccination course 4 weeks apart followed by their third dose 12 weeks later. Twenty vaccinated adult horses (10 each for WT or RG vaccine) received a single dose of a booster vaccination. Results The RG virus had high growth properties in embryonated chicken eggs. Unvaccinated yearlings responded poorly to the first vaccination, especially those that received the RG vaccine, but mounted better responses to the second and the third vaccinations, and maintained relatively high haemagglutination inhibition (HI) titres up to 28 weeks after the first vaccination. Vaccinated adult horses did not respond remarkably to the booster vaccination, but no horses showed titres below their pre-booster values even at 12 weeks after vaccination. The RG virus elicited immunogenicity in horses adequate for vaccine use. Main limitations No virus challenge study was performed. Conclusions The RG viruses are useful as an EI vaccine strain, and quick updates of an EI vaccine strain can be achieved by using RG techniques.
- Published
- 2021
95. Chimeric hPIV2/Corona-Spike Nasal Vaccine Robustly Protects the Upper and Lower Airways Against SARS-CoV-2
- Author
-
Ryoichi Ono, Seiya Yamayoshi, Yoshihiro Kawaoka, Tadashi Maemura, Masayuki Fukumura, Junpei Ohtsuka, Asami Eguchi, Maeda M, Mutsumi Ito, Yosky Kataoka, Tetsuya Nosaka, and Masaki Imai
- Subjects
biology ,business.industry ,Virology ,Viral vector ,Herd immunity ,Vaccination ,Viral envelope ,Animal Care Committees ,biology.protein ,Medicine ,Nasal administration ,Vector (molecular biology) ,Antibody ,business - Abstract
We developed an intranasal vaccine against SARS-CoV-2 using the replication-incompetent human parainfluenza virus type 2 (hPIV2) vector BC-PIV, which can deliver ectopic gene as stable RNA and ectopic protein on the envelope. BC-PIV expressing the full-length prefusion-stabilized spike gene of SARS-CoV-2, S2PM, possessed a corona-like viral envelope. Intranasal vaccination of mice with BC-PIV/S2PM induced high levels of neutralizing IgG and mucosal IgA antibodies against the spike protein. While BC-PIV showed hemagglutinating activity, BC-PIV/S2PM lacked such activity, in accordance with the presence of the massive spike protein on the viral surface. Furthermore, single-dose intranasal vaccination of hamsters with BC-PIV/S2PM completely protected the lungs from SARS-CoV-2 at 11 weeks post-immunization, and prime-boost vaccination conferred virtually complete protection of the nasal turbinates against SARS-CoV-2 challenge at 11 weeks post-priming. Thus, this chimeric hPIV2/spike intranasal vaccine is one of the strong candidates for an ultimate vaccine against SARS-CoV-2 to curtail virus transmission. Funding: This work was supported in part by Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology in Japan (17K19652, 20K21614), by a Research Program on Emerging and Re-emerging Infectious Diseases from the Japan Agency for Medical Research and Development (AMED) (JP19fk0108113), Mie University (for research institutes of excellence), Mie Prefecture, Junior Chamber International Yokkaichi, and MediciNova, Inc. Conflict of Interest: J.O., M.F., M.Im., R.O., S.Y., Y.Kaw., and T.N. are patent applicants for recombinant BCPIV vaccine against SARS-CoV-2. M.F. is a founder of BioComo, Inc., and J.O. is an employee of BioComo, Inc. J.O., M.F., M.M., and T.N. have shares of stock in Biocomo, Inc. M.M. is a scientific advisor of JEOL Ltd. T.N. is a scientific advisor of MediciNova, Inc. The other authors declare no competing interests. Ethical Approval: Recombinant DNA experiments with SARS-CoV-2 S gene fragments were approved by the Ministry of Education, Culture, Sports, Science and Technology in Japan (Approved No. 2019-728, 729; 2020-362, 373, 948). The animal studies were approved by the Animal Care Committees of Mie University(Approved No. 23-33) and the Animal Experiment Committee of the Institute of Medical Science, the University of Tokyo (Approved No. PA19-75), and all methods were performed under institutional regulations of animal experiments in accordance with the current national guidelines. Animal experiments using SARS-CoV-2 S gene fragments orSARS-CoV-2 were also approved by the Ministry of Education, Culture, Sports, Science and Technology in Japan (Approved No. 2019-728, 729; 2020-362, 373, 2020-948).
- Published
- 2021
96. Comparison of Rapid Antigen Tests for COVID-19
- Author
-
Masaki Imai, Eisuke Adachi, Michiko Koga, Kazuhiko Ikeuchi, Seiya Yamayoshi, Takato Nakamoto, Atsuhiro Yasuhara, Shinya Yamada, Mutsumi Ito, Kenji Maeda, Shin ichiro Hattori, Rie Baba, Yuko Sakai-Tagawa, Kiyoko Iwatsuki-Horimoto, Hidefumi Koh, Yuri Furusawa, Takahiro Fukui, Hiroshi Yotsuyanagi, Makoto Saito, Kensuke Fujita, Hiroaki Mitsuya, Norio Omagari, Fumimaro Ito, Ichiro Nakachi, Hiroyuki Nagai, Yoshihiro Kawaoka, Michiko Ujie, Osamu Akasaka, Takayuki Ogura, and Kei Yamamoto
- Subjects
0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,diagnosis ,Point-of-Care Systems ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,030106 microbiology ,lcsh:QR1-502 ,Real-Time Polymerase Chain Reaction ,Sensitivity and Specificity ,Article ,Virus ,lcsh:Microbiology ,COVID-19 Serological Testing ,Specimen Handling ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Virology ,Humans ,Medicine ,030212 general & internal medicine ,rapid antigen test ,Antigens, Viral ,False Negative Reactions ,Immunoassay ,medicine.diagnostic_test ,business.industry ,SARS-CoV-2 ,COVID-19 ,Infectious Diseases ,Real-time polymerase chain reaction ,Rapid antigen test ,business - Abstract
Reverse transcription-quantitative PCR (RT-qPCR)-based tests are widely used to diagnose coronavirus disease 2019 (COVID-19). As a result that these tests cannot be done in local clinics where RT-qPCR testing capability is lacking, rapid antigen tests (RATs) for COVID-19 based on lateral flow immunoassays are used for rapid diagnosis. However, their sensitivity compared with each other and with RT-qPCR and infectious virus isolation has not been examined. Here, we compared the sensitivity among four RATs by using severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) isolates and several types of COVID-19 patient specimens and compared their sensitivity with that of RT-qPCR and infectious virus isolation. Although the RATs read the samples containing large amounts of virus as positive, even the most sensitive RAT read the samples containing small amounts of virus as negative. Moreover, all RATs tested failed to detect viral antigens in several specimens from which the virus was isolated. The current RATs will likely miss some COVID-19 patients who are shedding infectious SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
97. Antibody titers against SARS-CoV-2 decline, but do not disappear for several months
- Author
-
Hiroaki Mitsuya, Takayuki Ogura, Shiho Chiba, Jurika Murakami, Michiko Koga, Junichi Ochi, Masanori Nojima, Ichiro Nakachi, Tetsuya Suzuki, Hiroyuki Nagai, Kenji Maeda, Norio Omagari, Kensuke Fujita, Makoto Saito, Osamu Akasaka, Rie Baba, Hideaki Nakajima, Yoshihiro Kawaoka, Hideaki Kato, Kazuma Yagi, Norio Sugaya, Kiyoko Iwatsuki-Horimoto, Eisuke Adachi, David J. Pattinson, Taiki Hamabata, Mutsumi Ito, Yusuke Miyazato, Keiko Mitamura, Hiroshi Yotsuyanagi, Shin ichiro Hattori, Atsuhiro Yasuhara, Morio Nakamura, Shinya Yamamoto, Moe Okuda, and Seiya Yamayoshi
- Subjects
lcsh:R5-920 ,Allergy ,biology ,business.industry ,010102 general mathematics ,Antibody titer ,General Medicine ,medicine.disease ,01 natural sciences ,03 medical and health sciences ,Titer ,0302 clinical medicine ,Ectodomain ,Immunology ,Cohort ,biology.protein ,Medicine ,030212 general & internal medicine ,0101 mathematics ,Antibody ,lcsh:Medicine (General) ,business ,Neutralizing antibody ,Pathogen ,Research Paper - Abstract
Background To develop an effective vaccine against a novel viral pathogen, it is important to understand the longitudinal antibody responses against its first infection. Here we performed a longitudinal study of antibody responses against SARS-CoV-2 in symptomatic patients. Methods Sequential blood samples were collected from 39 individuals at various timepoints between 0 and 154 days after onset. IgG or IgM titers to the receptor binding domain (RBD) of the S protein, the ectodomain of the S protein, and the N protein were determined by using an ELISA. Neutralizing antibody titers were measured by using a plaque reduction assay. Findings The IgG titers to the RBD of the S protein, the ectodomain of the S protein, and the N protein peaked at about 20 days after onset, gradually decreased thereafter, and were maintained for several months after onset. Extrapolation modeling analysis suggested that the IgG antibodies were maintained for this amount of time because the rate of reduction slowed after 30 days post-onset. IgM titers to the RBD decreased rapidly and disappeared in some individuals after 90 days post-onset. All patients, except one, possessed neutralizing antibodies against authentic SARS-CoV-2, which they retained at 90 days after onset. The highest antibody titers in patients with severe infections were higher than those in patients with mild or moderate infections, but the decrease in antibody titer in the severe infection cohort was more remarkable than that in the mild or moderate infection cohort. Interpretation Although the number of patients is limited, our results show that the antibody response against the first SARS-CoV-2 infection in symptomatic patients is typical of that observed in an acute viral infection. Funding The Japan Agency for Medical Research and Development and the National Institutes of Allergy and Infectious Diseases.
- Published
- 2020
98. Longitudinal antibody repertoire in 'mild' versus 'severe' COVID-19 patients reveals immune markers associated with disease severity and resolution
- Author
-
Makoto Saito, Surender Khurana, Atsuhiro Yasuhara, Seiya Yamayoshi, Shinya Yamada, Takayuki Ogura, Laura Klenow, Masaki Imai, Osamu Akasaka, Kiyoko Iwatsuki-Horimoto, Eisuke Adachi, Elizabeth M. Coyle, Yuko Sakai-Tagawa, Youri Lee, Hiroshi Yotsuyanagi, Supriya Ravichandran, Rie Baba, Ichiro Nakachi, Mutsumi Ito, Maki Kiso, Gabrielle Grubbs, Michiko Koga, and Yoshihiro Kawaoka
- Subjects
Antibody Affinity ,macromolecular substances ,Antibodies, Viral ,Severity of Illness Index ,Epitope ,Affinity maturation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antibody Repertoire ,Antigen ,Protein Domains ,Neutralization Tests ,Medicine ,Humans ,030212 general & internal medicine ,skin and connective tissue diseases ,Antigens, Viral ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,business.industry ,SARS-CoV-2 ,fungi ,COVID-19 ,SciAdv r-articles ,biochemical phenomena, metabolism, and nutrition ,Viral Load ,Immunoglobulin Class Switching ,body regions ,Hospitalization ,Coronavirus ,Kinetics ,HEK293 Cells ,Immunoglobulin class switching ,Immunoglobulin M ,Immunology ,Antibody Formation ,Spike Glycoprotein, Coronavirus ,biology.protein ,Cytokines ,Antibody ,business ,Biomarkers ,Protein Binding ,Research Article - Abstract
SARS-CoV-2–induced human antibody repertoire in patients with COVID-19 reveals immune markers of disease severity and recovery., Limited knowledge exists on immune markers associated with disease severity or recovery in patients with coronavirus disease 2019 (COVID-19). Here, we elucidated longitudinal evolution of SARS-CoV-2 antibody repertoire in patients with acute COVID-19. Differential kinetics was observed for immunoglobulin M (IgM)/IgG/IgA epitope diversity, antibody binding, and affinity maturation in “severe” versus “mild” COVID-19 patients. IgG profile demonstrated immunodominant antigenic sequences encompassing fusion peptide and receptor binding domain (RBD) in patients with mild COVID-19 who recovered early compared with “fatal” COVID-19 patients. In patients with severe COVID-19, high-titer IgA were observed, primarily against RBD, especially in patients who succumbed to SARS-CoV-2 infection. The patients with mild COVID-19 showed marked increase in antibody affinity maturation to prefusion SARS-CoV-2 spike that associated with faster recovery from COVID-19. This study revealed antibody markers associated with disease severity and resolution of clinical disease that could inform development and evaluation of effective immune-based countermeasures against COVID-19.
- Published
- 2020
99. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development
- Author
-
Yuri Furusawa, Noriko Kinoshita, Noriko Nakajima, Tadaki Suzuki, Aaron Balogh, Tokiko Watanabe, Peter Halfmann, Samantha Loeber, Moe Okuda, Mutsumi Ito, Seiya Yamayoshi, Makoto Takeda, Lizheng Guan, Florian Krammer, Shufang Fan, Kenta Takahashi, Kiyoko Iwatsuki-Horimoto, Masaki Imai, Hiroshi Ueki, Tammy Armbrust, Masato Hatta, Shin-ichiro Hattori, Tiago J. S. Lopes, Atsuhiro Yasuhara, Shiho Chiba, Yuko Sakai-Tagawa, Norio Ohmagari, Shinya Yamada, Hiroaki Mitsuya, Makoto Kuroda, Michiko Ujie, Yoshihiro Kawaoka, Maki Kiso, and Kosuke Takada
- Subjects
0301 basic medicine ,viruses ,medicine.disease_cause ,Antibodies, Viral ,Virus Replication ,Pathogenesis ,0302 clinical medicine ,Cricetinae ,Chlorocebus aethiops ,Neutralizing antibody ,skin and connective tissue diseases ,Lung ,Coronavirus ,Multidisciplinary ,biology ,virus diseases ,respiratory system ,Biological Sciences ,medicine.anatomical_structure ,Ribonucleoproteins ,Coronavirus Infections ,Pneumonia, Viral ,Hamster ,Lung injury ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Betacoronavirus ,Viral Proteins ,medicine ,Animals ,Humans ,Pandemics ,Vero Cells ,COVID-19 Serotherapy ,Mesocricetus ,business.industry ,SARS-CoV-2 ,fungi ,Immunization, Passive ,COVID-19 ,biology.organism_classification ,Virology ,Antibodies, Neutralizing ,infection ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,business ,Syrian hamsters ,countermeasure ,030217 neurology & neurosurgery - Abstract
Significance Since SARS-CoV-2 emerged in China, it has spread rapidly around the world. Effective vaccines and therapeutics for SARS-CoV-2−induced disease (coronavirus disease 2019;COVID-19) are urgently needed. We found that SARS-CoV-2 isolates replicate efficiently in the lungs of Syrian hamsters and cause severe pathological lesions in the lungs of these animals similar to commonly reported imaging features of COVID-19 patients with pneumonia. SARS-CoV-2−infected hamsters mounted neutralizing antibody responses and were protected against rechallenge with SARS-CoV-2. Moreover, passive transfer of convalescent serum to naïve hamsters inhibited virus replication in their lungs. Syrian hamsters are a useful small animal model for the evaluation of vaccines, immunotherapies, and antiviral drugs., At the end of 2019, a novel coronavirus (severe acute respiratory syndrome coronavirus 2; SARS-CoV-2) was detected in Wuhan, China, that spread rapidly around the world, with severe consequences for human health and the global economy. Here, we assessed the replicative ability and pathogenesis of SARS-CoV-2 isolates in Syrian hamsters. SARS-CoV-2 isolates replicated efficiently in the lungs of hamsters, causing severe pathological lung lesions following intranasal infection. In addition, microcomputed tomographic imaging revealed severe lung injury that shared characteristics with SARS-CoV-2−infected human lung, including severe, bilateral, peripherally distributed, multilobular ground glass opacity, and regions of lung consolidation. SARS-CoV-2−infected hamsters mounted neutralizing antibody responses and were protected against subsequent rechallenge with SARS-CoV-2. Moreover, passive transfer of convalescent serum to naïve hamsters efficiently suppressed the replication of the virus in the lungs even when the serum was administrated 2 d postinfection of the serum-treated hamsters. Collectively, these findings demonstrate that this Syrian hamster model will be useful for understanding SARS-CoV-2 pathogenesis and testing vaccines and antiviral drugs.
- Published
- 2020
100. Rapid and accurate detection of novel coronavirus SARS-CoV-2 using CRISPR-Cas3
- Author
-
Hiroshi Yotsuyanagi, Masayuki Yamamoto, Satomi Shibumura, Kohei Takeshita, Seiya Yamayoshi, Yuko Yamauchi, Yoshihiro Kawaoka, Tomoji Mashimo, and Kazuto Yoshimi
- Subjects
Dna cleavage ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine ,CRISPR ,In patient ,Severe acute respiratory syndrome coronavirus ,Biology ,medicine.disease_cause ,Diagnostic tools ,Virology ,Virus ,Coronavirus - Abstract
Novel coronavirus SARS-CoV-2 outbreaks have rapidly spread to multiple countries, highlighting the urgent necessity for fast, sensitive, and specific diagnostic tools for virus surveillance. Here, the previously unknown collateral single-stranded DNA cleavage we observed with type I CRISPR-Cas3 highlights its potential for development as a Cas3-mediated rapid (within 40 min), low-cost, instrument-free detection method for SARS-CoV-2. This Cas3-based assay is comparable with Cas12- and real-time reverse-transcriptase PCR-based assays in its speed and sensitivity, but offers greater specificity for single-base-pair discrimination while negating the need for highly trained operators. These findings support the use of CRISPR diagnostics for point-of-care testing in patients with suspected SARS-CoV-2 infections.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.