43 results on '"Wildum, S"'
Search Results
2. Tocilizumab in patients hospitalised with COVID-19 pneumonia: Efficacy, safety, viral clearance, and antibody response from a randomised controlled trial (COVACTA)
- Author
-
Rosas, I, Bräu, N, Waters, M, Go, R, Malhotra, A, Hunter, B, Bhagani, S, Skiest, D, Savic, S, Douglas, I, Garcia-Diaz, J, Aziz, M, Cooper, N, Youngstein, T, Sorbo, L, Zerda, D, Ustianowski, A, Gracian, A, Blyth, K, Carratalà, J, François, B, Benfield, T, Haslem, D, Bonfanti, P, van der Leest, C, Rohatgi, N, Wiese, L, Luyt, C, Bauer, R, Cai, F, Lee, I, Matharu, B, Metcalf, L, Wildum, S, Graham, E, Tsai, L, Bao, M, Rosas, Ivan O, Bräu, Norbert, Waters, Michael, Go, Ronaldo C, Malhotra, Atul, Hunter, Bradley D, Bhagani, Sanjay, Skiest, Daniel, Savic, Sinisa, Douglas, Ivor S, Garcia-Diaz, Julia, Aziz, Mariam S, Cooper, Nichola, Youngstein, Taryn, Sorbo, Lorenzo Del, Zerda, David J De La, Ustianowski, Andrew, Gracian, Antonio Cubillo, Blyth, Kevin G, Carratalà, Jordi, François, Bruno, Benfield, Thomas, Haslem, Derrick, Bonfanti, Paolo, van der Leest, Cor H, Rohatgi, Nidhi, Wiese, Lothar, Luyt, Charles Edouard, Bauer, Rebecca N, Cai, Fang, Lee, Ivan T, Matharu, Balpreet, Metcalf, Louis, Wildum, Steffen, Graham, Emily, Tsai, Larry, Bao, Min, Rosas, I, Bräu, N, Waters, M, Go, R, Malhotra, A, Hunter, B, Bhagani, S, Skiest, D, Savic, S, Douglas, I, Garcia-Diaz, J, Aziz, M, Cooper, N, Youngstein, T, Sorbo, L, Zerda, D, Ustianowski, A, Gracian, A, Blyth, K, Carratalà, J, François, B, Benfield, T, Haslem, D, Bonfanti, P, van der Leest, C, Rohatgi, N, Wiese, L, Luyt, C, Bauer, R, Cai, F, Lee, I, Matharu, B, Metcalf, L, Wildum, S, Graham, E, Tsai, L, Bao, M, Rosas, Ivan O, Bräu, Norbert, Waters, Michael, Go, Ronaldo C, Malhotra, Atul, Hunter, Bradley D, Bhagani, Sanjay, Skiest, Daniel, Savic, Sinisa, Douglas, Ivor S, Garcia-Diaz, Julia, Aziz, Mariam S, Cooper, Nichola, Youngstein, Taryn, Sorbo, Lorenzo Del, Zerda, David J De La, Ustianowski, Andrew, Gracian, Antonio Cubillo, Blyth, Kevin G, Carratalà, Jordi, François, Bruno, Benfield, Thomas, Haslem, Derrick, Bonfanti, Paolo, van der Leest, Cor H, Rohatgi, Nidhi, Wiese, Lothar, Luyt, Charles Edouard, Bauer, Rebecca N, Cai, Fang, Lee, Ivan T, Matharu, Balpreet, Metcalf, Louis, Wildum, Steffen, Graham, Emily, Tsai, Larry, and Bao, Min
- Abstract
Background: In COVACTA, a randomised, placebo-controlled trial in patients hospitalised with coronavirus disease-19 (COVID-19), tocilizumab did not improve 28-day mortality, but shortened hospital and intensive care unit stay. Longer-term effects of tocilizumab in patients with COVID-19 are unknown. Therefore, the efficacy and safety of tocilizumab in COVID-19 beyond day 28 and its impact on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) clearance and antibody response in COVACTA were investigated. Methods: Adults in Europe and North America hospitalised with COVID-19 (N = 452) between April 3, 2020 and May 28, 2020 were randomly assigned (2:1) to double-blind intravenous tocilizumab or placebo and assessed for efficacy and safety through day 60. Assessments included mortality, time to hospital discharge, SARS-CoV-2 viral load in nasopharyngeal swab and serum samples, and neutralising anti-SARS-CoV-2 antibodies in serum. ClinicalTrials.gov registration: NCT04320615. Findings: By day 60, 24·5% (72/294) of patients in the tocilizumab arm and 25·0% (36/144) in the placebo arm died (weighted difference -0·5% [95% CI -9·1 to 8·0]), and 67·0% (197/294) in the tocilizumab arm and 63·9% (92/144) in the placebo arm were discharged from the hospital. Serious infections occurred in 24·1% (71/295) of patients in the tocilizumab arm and 29·4% (42/143) in the placebo arm. Median time to negative reverse transcriptase-quantitative polymerase chain reaction result in nasopharyngeal/oropharyngeal samples was 15·0 days (95% CI 14·0 to 21·0) in the tocilizumab arm and 21·0 days (95% CI 14·0 to 28·0) in the placebo arm. All tested patients had positive test results for neutralising anti-SARS-CoV-2 antibodies at day 60. Interpretation: There was no mortality benefit with tocilizumab through day 60. Tocilizumab did not impair viral clearance or host immune response, and no new safety signals were observed. Future investigations may explore potential biomarkers to
- Published
- 2022
3. Assessing the fitness of a dual-antiviral drug resistant human influenza virus in the ferret model
- Author
-
Stannard, HL, Mifsud, EJ, Wildum, S, Brown, SK, Koszalka, P, Shishido, T, Kojima, S, Omoto, S, Baba, K, Kuhlbusch, K, Hurt, AC, Barr, IG, Stannard, HL, Mifsud, EJ, Wildum, S, Brown, SK, Koszalka, P, Shishido, T, Kojima, S, Omoto, S, Baba, K, Kuhlbusch, K, Hurt, AC, and Barr, IG
- Abstract
Influenza antivirals are important tools in our fight against annual influenza epidemics and future influenza pandemics. Combinations of antivirals may reduce the likelihood of drug resistance and improve clinical outcomes. Previously, two hospitalised immunocompromised influenza patients, who received a combination of a neuraminidase inhibitor and baloxavir marboxil, shed influenza viruses resistant to both drugs. Here-in, the replicative fitness of one of these A(H1N1)pdm09 virus isolates with dual resistance mutations (NA-H275Y and PA-I38T) was similar to wild type virus (WT) in vitro, but reduced in the upper respiratory tracts of challenged ferrets. The dual-mutant virus transmitted well between ferrets in an airborne transmission model, but was outcompeted by the WT when the two viruses were co-administered. These results indicate the dual-mutant virus had a moderate loss of viral fitness compared to the WT virus, suggesting that while person-to-person transmission of the dual-resistant virus may be possible, widespread community transmission is unlikely.
- Published
- 2022
4. Evaluating the fitness of PA/I38T-substituted influenza A viruses with reduced baloxavir susceptibility in a competitive mixtures ferret model
- Author
-
Lowen, AC, Lee, LYY, Zhou, J, Koszalka, P, Frise, R, Farrukee, R, Baba, K, Miah, S, Shishido, T, Galiano, M, Hashimoto, T, Omoto, S, Uehara, T, Mifsud, EJ, Collinson, N, Kuhlbusch, K, Clinch, B, Wildum, S, Barclay, WS, Hurt, AC, Lowen, AC, Lee, LYY, Zhou, J, Koszalka, P, Frise, R, Farrukee, R, Baba, K, Miah, S, Shishido, T, Galiano, M, Hashimoto, T, Omoto, S, Uehara, T, Mifsud, EJ, Collinson, N, Kuhlbusch, K, Clinch, B, Wildum, S, Barclay, WS, and Hurt, AC
- Abstract
Baloxavir is approved in several countries for the treatment of uncomplicated influenza in otherwise-healthy and high-risk patients. Treatment-emergent viruses with reduced susceptibility to baloxavir have been detected in clinical trials, but the likelihood of widespread occurrence depends on replication capacity and onward transmission. We evaluated the fitness of A/H3N2 and A/H1N1pdm09 viruses with the polymerase acidic (PA) I38T-variant conferring reduced susceptibility to baloxavir relative to wild-type (WT) viruses, using a competitive mixture ferret model, recombinant viruses and patient-derived virus isolates. The A/H3N2 PA/I38T virus showed a reduction in within-host fitness but comparable between-host fitness to the WT virus, while the A/H1N1pdm09 PA/I38T virus had broadly similar within-host fitness but substantially lower between-host fitness. Although PA/I38T viruses replicate and transmit between ferrets, our data suggest that viruses with this amino acid substitution have lower fitness relative to WT and this relative fitness cost was greater in A/H1N1pdm09 viruses than in A/H3N2 viruses.
- Published
- 2021
5. Baloxavir treatment of ferrets infected with influenza A(H1N1)pdm09 virus reduces onward transmission
- Author
-
Lowen, AC, Lee, LYY, Zhou, J, Frise, R, Goldhill, DH, Koszalka, P, Mifsud, EJ, Baba, K, Noda, T, Ando, Y, Sato, K, Yuki, A-I, Shishido, T, Uehara, T, Wildum, S, Zwanziger, E, Collinson, N, Kuhlbusch, K, Clinch, B, Hurt, AC, Barclay, WS, Lowen, AC, Lee, LYY, Zhou, J, Frise, R, Goldhill, DH, Koszalka, P, Mifsud, EJ, Baba, K, Noda, T, Ando, Y, Sato, K, Yuki, A-I, Shishido, T, Uehara, T, Wildum, S, Zwanziger, E, Collinson, N, Kuhlbusch, K, Clinch, B, Hurt, AC, and Barclay, WS
- Abstract
Influenza viruses cause seasonal outbreaks and pose a continuous pandemic threat. Although vaccines are available for influenza control, their efficacy varies each season and a vaccine for a novel pandemic virus manufactured using current technology will not be available fast enough to mitigate the effect of the first pandemic wave. Antivirals can be effective against many different influenza viruses but have not thus far been used extensively for outbreak control. Baloxavir, a recently licensed antiviral drug that targets the influenza virus endonuclease, has been shown to reduce virus shedding more effectively than oseltamivir, a widely used neuraminidase inhibitor drug. Thus it is possible that treatment with baloxavir might also interrupt onward virus transmission. To test this, we utilized the ferret model, which is the most commonly used animal model to study influenza virus transmission. We established a subcutaneous baloxavir administration method in ferrets which achieved similar pharmacokinetics to the approved human oral dose. Transmission studies were then conducted in two different locations with different experimental setups to compare the onward transmission of A(H1N1)pdm09 virus from infected ferrets treated with baloxavir, oseltamivir or placebo to naïve sentinel ferrets exposed either indirectly in adjacent cages or directly by co-housing. We found that baloxavir treatment reduced infectious viral shedding in the upper respiratory tract of ferrets compared to placebo, and reduced the frequency of transmission amongst sentinels in both experimental setups, even when treatment was delayed until 2 days post-infection. In contrast, oseltamivir treatment did not substantially affect viral shedding or transmission compared to placebo. We did not detect the emergence of baloxavir-resistant variants in treated animals or in untreated sentinels. Our results support the concept that antivirals which decrease viral shedding could also reduce influenza transmi
- Published
- 2020
6. Machine-learning based patient classification using Hepatitis B virus full-length genome quasispecies from Asian and European cohorts
- Author
-
Mueller-Breckenridge, A.J. (Alan J.), Garcia-Alcalde, F. (Fernando), Wildum, S. (Steffen), Smits, S.L. (Saskia), Man, R.A. (Robert) de, Campenhout, M.J.H. (Margo) van, Brouwer, W.P. (Willem), Niu, J. (Jianjun), Young, J.A.T. (John A T), Najera, I. (Isabel), Zhu, L. (Lina), Wu, D. (Daitze), Racek, T. (Tomas), Hundie, G.B. (Gadissa Bedada), Lin, Y. (Yong), Boucher, C.A.B. (Charles), Vijver, D.A.M.C. (David) van de, Haagmans, B.L. (Bart), Mueller-Breckenridge, A.J. (Alan J.), Garcia-Alcalde, F. (Fernando), Wildum, S. (Steffen), Smits, S.L. (Saskia), Man, R.A. (Robert) de, Campenhout, M.J.H. (Margo) van, Brouwer, W.P. (Willem), Niu, J. (Jianjun), Young, J.A.T. (John A T), Najera, I. (Isabel), Zhu, L. (Lina), Wu, D. (Daitze), Racek, T. (Tomas), Hundie, G.B. (Gadissa Bedada), Lin, Y. (Yong), Boucher, C.A.B. (Charles), Vijver, D.A.M.C. (David) van de, and Haagmans, B.L. (Bart)
- Abstract
Chronic infection with Hepatitis B virus (HBV) is a major risk factor for the development of advanced liver disease including fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The relative contribution of virological factors to disease progression has not been fully defined and tools aiding the deconvolution of complex patient virus profiles is an unmet clinical need. Varia
- Published
- 2019
- Full Text
- View/download PDF
7. Machine-learning based patient classification using Hepatitis B virus full-length genome quasispecies from Asian and European cohorts
- Author
-
Mueller-Breckenridge, AJ, Garcia-Alcalde, F, Wildum, S, Smits, Saskia, de Man, Rob, van Campenhout, Margo, Brouwer, Willem Pieter, Niu, JJ, Young, JAT, Najera, I, Zhu, LN, Wu, D, Racek, T, Hundie, Gadissa, Lin, Y, Boucher, Charles, van de Vijver, David, Haagmans, Bart, Mueller-Breckenridge, AJ, Garcia-Alcalde, F, Wildum, S, Smits, Saskia, de Man, Rob, van Campenhout, Margo, Brouwer, Willem Pieter, Niu, JJ, Young, JAT, Najera, I, Zhu, LN, Wu, D, Racek, T, Hundie, Gadissa, Lin, Y, Boucher, Charles, van de Vijver, David, and Haagmans, Bart
- Published
- 2019
8. Antiviral efficacy of a novel hepatitis B virus expression inhibitor in combination with entecavir and wIFN-alpha in woodchucks model of chronic hepatitis B
- Author
-
Wildum, S., primary, Steiner, G., additional, Luangsay, S., additional, Suresh, M., additional, Korolowicz, K., additional, Balarezo, M., additional, Yon, C., additional, Murreddu, M., additional, Hong, X., additional, Kallakury, B., additional, Tucker, R., additional, Mueller, H., additional, Menne, S., additional, Young, J., additional, and Javanbakht, H., additional
- Published
- 2017
- Full Text
- View/download PDF
9. Tocilizumab in patients hospitalised with COVID-19 pneumonia: Efficacy, safety, viral clearance, and antibody response from a randomised controlled trial (COVACTA)
- Author
-
Ivan O. Rosas, Norbert Bräu, Michael Waters, Ronaldo C. Go, Atul Malhotra, Bradley D. Hunter, Sanjay Bhagani, Daniel Skiest, Sinisa Savic, Ivor S. Douglas, Julia Garcia-Diaz, Mariam S. Aziz, Nichola Cooper, Taryn Youngstein, Lorenzo Del Sorbo, David J. De La Zerda, Andrew Ustianowski, Antonio Cubillo Gracian, Kevin G. Blyth, Jordi Carratalà, Bruno François, Thomas Benfield, Derrick Haslem, Paolo Bonfanti, Cor H. van der Leest, Nidhi Rohatgi, Lothar Wiese, Charles Edouard Luyt, Rebecca N. Bauer, Fang Cai, Ivan T. Lee, Balpreet Matharu, Louis Metcalf, Steffen Wildum, Emily Graham, Larry Tsai, Min Bao, Rosas, I, Bräu, N, Waters, M, Go, R, Malhotra, A, Hunter, B, Bhagani, S, Skiest, D, Savic, S, Douglas, I, Garcia-Diaz, J, Aziz, M, Cooper, N, Youngstein, T, Sorbo, L, Zerda, D, Ustianowski, A, Gracian, A, Blyth, K, Carratalà, J, François, B, Benfield, T, Haslem, D, Bonfanti, P, van der Leest, C, Rohatgi, N, Wiese, L, Luyt, C, Bauer, R, Cai, F, Lee, I, Matharu, B, Metcalf, L, Wildum, S, Graham, E, Tsai, L, and Bao, M
- Subjects
Randomised controlled trial ,Coronavirus disease 2019 ,Interleukin-6 ,Prevention ,Clinical Trials and Supportive Activities ,COVID-19 ,Evaluation of treatments and therapeutic interventions ,General Medicine ,Tocilizumab ,Severe acute respiratory syndrome coronavirus-2 ,Vaccine Related ,Infectious Diseases ,Emerging Infectious Diseases ,Good Health and Well Being ,Clinical Research ,6.1 Pharmaceuticals ,Pneumonia & Influenza ,Viral load ,skin and connective tissue diseases ,Infection ,Lung - Abstract
Background: \ud In COVACTA, a randomised, placebo-controlled trial in patients hospitalised with coronavirus disease-19 (COVID-19), tocilizumab did not improve 28-day mortality, but shortened hospital and intensive care unit stay. Longer-term effects of tocilizumab in patients with COVID-19 are unknown. Therefore, the efficacy and safety of tocilizumab in COVID-19 beyond day 28 and its impact on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) clearance and antibody response in COVACTA were investigated.\ud \ud Methods: \ud Adults in Europe and North America hospitalised with COVID-19 (N = 452) between April 3, 2020 and May 28, 2020 were randomly assigned (2:1) to double-blind intravenous tocilizumab or placebo and assessed for efficacy and safety through day 60. Assessments included mortality, time to hospital discharge, SARS-CoV-2 viral load in nasopharyngeal swab and serum samples, and neutralising anti-SARS-CoV-2 antibodies in serum. ClinicalTrials.gov registration: NCT04320615.\ud \ud Findings: \ud By day 60, 24·5% (72/294) of patients in the tocilizumab arm and 25·0% (36/144) in the placebo arm died (weighted difference –0·5% [95% CI –9·1 to 8·0]), and 67·0% (197/294) in the tocilizumab arm and 63·9% (92/144) in the placebo arm were discharged from the hospital. Serious infections occurred in 24·1% (71/295) of patients in the tocilizumab arm and 29·4% (42/143) in the placebo arm. Median time to negative reverse transcriptase–quantitative polymerase chain reaction result in nasopharyngeal/oropharyngeal samples was 15·0 days (95% CI 14·0 to 21·0) in the tocilizumab arm and 21·0 days (95% CI 14·0 to 28·0) in the placebo arm. All tested patients had positive test results for neutralising anti–SARS-CoV-2 antibodies at day 60.\ud \ud Interpretation: \ud There was no mortality benefit with tocilizumab through day 60. Tocilizumab did not impair viral clearance or host immune response, and no new safety signals were observed. Future investigations may explore potential biomarkers to optimize patient selection for tocilizumab treatment and combination therapy with other treatments.\ud \ud Funding: \ud F. Hoffmann-La Roche Ltd and the US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, under OT number HHSO100201800036C.
- Published
- 2021
10. Comparison of influenza viral load in nasopharyngeal and midturbinate swabs.
- Author
-
Bernasconi C, Katugampola L, and Wildum S
- Subjects
- Humans, Viral Load, Serologic Tests, Nasopharynx, Influenza, Human diagnosis
- Abstract
Different specimen types are used for influenza diagnosis but comparative data for viral loads from different swab types are limited. We compared influenza viral loads (determined by quantitative reverse transcription polymerase chain reaction [RT-PCR]) in 93 paired midturbinate and nasopharyngeal swab aliquots from influenza infected patients enrolled in a phase 3 randomized-controlled study with the objective of maximizing the number of swabs available for sequence analysis. Midturbinate swabs yielded a 53% lower viral load versus nasopharyngeal swabs, and this difference was similar for influenza A and B. These data suggest that nasopharyngeal swabs might be preferred in diagnostic settings when obtaining higher viral load is important., Competing Interests: C. B. has received consulting fees as a contractor for F. Hoffmann‐La Roche Ltd. during the execution of this work. L. K. and S. W. are employees of, and own stocks in, F. Hoffmann‐La Roche Ltd., (© 2024 F.Hoffmann‐La Roche Ltd. Influenza and Other Respiratory Viruses published by John Wiley & Sons Ltd.)
- Published
- 2024
- Full Text
- View/download PDF
11. Oral bemnifosbuvir (AT-527) vs placebo in patients with mild-to-moderate COVID-19 in an outpatient setting (MORNINGSKY).
- Author
-
Horga A, Saenz R, Yilmaz G, Simón-Campos A, Pietropaolo K, Stubbings WJ, Collinson N, Ishak L, Zrinscak B, Belanger B, Granier C, Lin K, C Hurt A, Zhou XJ, Wildum S, and Hammond J
- Abstract
Aim: This phase III study assessed the efficacy/safety/antiviral activity/pharmacokinetics of bemnifosbuvir, a novel, oral nucleotide analog to treat COVID-19. Patients & methods: Outpatient adults/adolescents with mild-to-moderate COVID-19 were randomized 2:1 to bemnifosbuvir/placebo. Time to symptom alleviation/improvement (primary outcome), risk of hospitalization/death, viral load and safety were evaluated. Results: Although the study was discontinued prematurely and did not meet its primary end point, bemnifosbuvir treatment resulted in fewer hospitalizations (71% relative risk reduction), COVID-19-related medically attended hospital visits, and COVID-19-related complications compared with placebo. No reduction in viral load was observed. The proportion of patients with adverse events was similar; no deaths occurred. Conclusion: Bemnifosbuvir showed hospitalization reduction in patients with variable disease progression risk and was well tolerated. Clinical Trial Registration : NCT04889040 (ClinicalTrials.gov)., Competing Interests: Competing interests disclosure A Horga, K Pietropaolo, L Ishak and B Belanger are employees of Atea. R Saenz is an employee and owns stock for Genentech (a member of the Roche group). A Simón-Campos is a speaker and advisory board member for Pfizer, AstraZeneca, Roche and Regeneron. WJ Stubbings, N Collinson, C Granier, AC Hurt and S Wildum are employees and shareholders of F. Hoffman-La-Roche Ltd. B Zrinscak is an employee of F. Hoffman-La-Roche Ltd. K Lin is a former employee of Atea. X-J Zhou and J Hammond are employees and shareholders of Atea. The authors have no other competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript apart from those disclosed., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
12. A Phase 2 Randomized Trial Evaluating the Antiviral Activity and Safety of the Direct-Acting Antiviral Bemnifosbuvir in Ambulatory Patients with Mild or Moderate COVID-19 (MOONSONG Study).
- Author
-
Boffito M, Dolan E, Singh K, Holmes W, Wildum S, Horga A, Pietropaolo K, Zhou XJ, Clinch B, Collinson N, and Ukachukwu V
- Subjects
- Humans, Antiviral Agents adverse effects, SARS-CoV-2, Treatment Outcome, COVID-19, Hepatitis C, Chronic
- Abstract
Bemnifosbuvir is an oral antiviral drug with a dual mechanism of action targeting viral RNA polymerase, with in vitro activity against SARS-CoV-2. We conducted a phase 2, double-blind study evaluating the antiviral activity, safety, efficacy, and pharmacokinetics of bemnifosbuvir in ambulatory patients with mild/moderate COVID-19. Patients were randomized 1:1 to bemnifosbuvir 550 mg or placebo (cohort A) and 3:1 to bemnifosbuvir 1,100 mg or placebo (cohort B); all doses were given twice daily for 5 days. The primary endpoint was a change from baseline in the amount of nasopharyngeal SARS-CoV-2 viral RNA by reverse transcription PCR (RT-PCR). The modified intent-to-treat infected population comprised 100 patients (bemnifosbuvir 550 mg, n = 30; bemnifosbuvir 1,100 mg, n = 30; cohort A placebo, n = 30; cohort B placebo, n = 10). The primary endpoint was not met: the difference in viral RNA adjusted means at day 7 was -0.25 log
10 copies/mL between bemnifosbuvir 550 mg and cohort A placebo (80% confidence interval [CI], -0.66 to 0.16; P = 0.4260), and -0.08 log10 copies/mL between bemnifosbuvir 1,100 mg and pooled placebo (80% CI, -0.48 to 0.33; P = 0.8083). Bemnifosbuvir 550 mg was well tolerated. Incidence of nausea and vomiting was higher with bemnifosbuvir 1,100 mg (10.0% and 16.7% of patients, respectively) than pooled placebo (2.5% nausea, 2.5% vomiting). In the primary analysis, bemnifosbuvir did not show meaningful antiviral activity on nasopharyngeal viral load as measured by RT-PCR compared with placebo in patients with mild/moderate COVID-19. The trial is registered at ClinicalTrials.gov under registration number NCT04709835. IMPORTANCE COVID-19 continues to be a major global public health challenge, and there remains a need for effective and convenient direct-acting antivirals that can be administered outside health care settings. Bemnifosbuvir is an oral antiviral with a dual mechanism of action and potent in vitro activity against SARS-CoV-2. In this study, we evaluated the antiviral activity, safety, efficacy, and pharmacokinetics of bemnifosbuvir in ambulatory patients with mild/moderate COVID-19. In the primary analysis, bemnifosbuvir did not show meaningful antiviral activity compared with placebo as assessed by nasopharyngeal viral loads. The negative predictive value of nasopharyngeal viral load reduction for clinical outcomes in COVID-19 is currently unclear, and further evaluation of bemnifosbuvir for COVID-19 may be warranted despite the findings observed in this study., Competing Interests: The authors declare a conflict of interest. This work was supported by F. Hoffmann-La Roche Ltd and Atea Pharmaceuticals. The study was designed and co-funded by F. Hoffmann-La Roche Ltd and Atea Pharmaceuticals. Third-party medical writing assistance was provided by Beth de Klerk, of Ashfield MedComms, an Inizio company, and was funded by F. Hoffmann-La Roche Ltd. M.B. has been an advisor for and received travel and research grants (to the organization) from Janssen, Roche, ViiV, Bristol-Myers Squibb, Pfizer, Merck Sharp & Dohme, Gilead, Mylan, Cipla, Teva, GSK, Novavax, and Valneva. E.D.: No conflict. K.S.: No conflict. W.H., B.C., N.C., and V.U. are employees of Roche Products Ltd and hold shares of F. Hoffmann-La Roche Ltd. S.W. is an employee and holds shares of F. Hoffmann-La Roche Ltd. A.H., K.P., and X.-J.Z. are employees of Atea Pharmaceuticals.- Published
- 2023
- Full Text
- View/download PDF
13. Prognostic value of severe acute respiratory syndrome coronavirus-2 viral load and antibodies in patients hospitalized with COVID-19.
- Author
-
Bauer RN, Teterina A, Shivram H, McBride J, Rosenberger CM, Cai F, Bao M, Tsai L, Gordon O, Lee IT, Wallin JJ, Porter D, Juneja K, Camus G, Rosas IO, and Wildum S
- Subjects
- Humans, SARS-CoV-2, Prognosis, Viral Load, Lung, Antibodies, Viral, COVID-19
- Abstract
Observational studies have identified the potential prognostic value for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) viral load and anti-SARS-CoV-2 antibodies in coronavirus disease 2019 (COVID-19). However, viral load in nasopharyngeal (NP) swabs produced inconsistent results in prognostic analyses, and the prognostic value of viral load or antibodies has not been confirmed in large clinical trials. COVACTA and REMDACTA were double-blind, randomized, controlled trials with a combined enrollment of 1078 patients hospitalized with COVID-19 treated with tocilizumab or placebo in COVACTA or tocilizumab plus remdesivir or placebo plus remdesivir in REMDACTA. We assessed the potential prognostic value of NP and serum SARS-CoV-2 viral load and serum anti-SARS-CoV-2 antibodies at baseline as biomarkers for clinical outcomes in patients enrolled in these trials. In adjusted Cox proportional hazard models, serum viral load was a more reliable predictor of clinical outcomes than NP viral load; high serum viral load was associated with higher risk for death and mechanical ventilation/death and lower likelihood of hospital discharge (high vs. negative viral load hazard ratios [95% confidence interval {CI}] were 2.87 [1.57-5.25], 3.86 [2.23-6.68], and 0.23 [0.14-0.36], respectively, in COVACTA and 8.11 [2.95-22.26], 10.29 [4.5-23.55], and 0.21 [0.15-0.29], respectively, in REMDACTA) and high serum viral load correlated with levels of inflammatory cytokines and lung damage biomarkers. High anti-SARS-CoV-2 spike protein antibody (ACOV2S) levels were associated with higher likelihood of hospital discharge (high vs. below the limit of quantification hazard ratios [95% CI] were 2.55 [1.59-4.08] for COVACTA and 1.54 [1.13-2.09] for REMDACTA). These results support the role of baseline SARS-CoV-2 serum viral load and ACOV2S antibody titers in predicting clinical outcomes for patients hospitalized with COVID-19., (© 2023 F. Hoffmann-La Roche Ltd and The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2023
- Full Text
- View/download PDF
14. Investigating the transmission of baloxavir-resistant influenza viruses from treated index patients to untreated household contacts in the BLOCKSTONE study.
- Author
-
Harding J, Bernasconi C, Williams S, Wildum S, Kinoshita M, Uehara T, and Hurt AC
- Subjects
- Humans, Drug Resistance, Viral, Pyridines pharmacology, Pyridines therapeutic use, Thiepins pharmacology, Thiepins therapeutic use, Clinical Trials, Phase III as Topic, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Influenza, Human drug therapy, Influenza, Human epidemiology
- Abstract
In a post-hoc analysis of the phase 3 BLOCKSTONE study (JapicCTI-184180), we investigated household transmission of baloxavir-resistant (PA/I38X) influenza viruses. Using baloxavir resistance rates from prior clinical trials and the rate of influenza transmission observed in the study, the predicted number of PA/I38X transmission events was 4.8, assuming wild type and PA/I38X viruses were equally transmissible. However, no PA/I38X viruses were observed. These results suggest a low potential for baloxavir-resistant influenza virus transmission from treated to untreated individuals, potentially due to reduced viral/transmission fitness for PA/I38X viruses and/or low viral titres at the time when resistant viruses arise., (© 2023 F. Hoffmann-La Roche Ltd. Influenza and Other Respiratory Viruses published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
15. Assessing the fitness of a dual-antiviral drug resistant human influenza virus in the ferret model.
- Author
-
Stannard HL, Mifsud EJ, Wildum S, Brown SK, Koszalka P, Shishido T, Kojima S, Omoto S, Baba K, Kuhlbusch K, Hurt AC, and Barr IG
- Subjects
- Animals, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, Drug Resistance, Viral genetics, Ferrets, Humans, Neuraminidase genetics, Virus Replication genetics, Influenza A Virus, H1N1 Subtype genetics, Influenza, Human drug therapy
- Abstract
Influenza antivirals are important tools in our fight against annual influenza epidemics and future influenza pandemics. Combinations of antivirals may reduce the likelihood of drug resistance and improve clinical outcomes. Previously, two hospitalised immunocompromised influenza patients, who received a combination of a neuraminidase inhibitor and baloxavir marboxil, shed influenza viruses resistant to both drugs. Here-in, the replicative fitness of one of these A(H1N1)pdm09 virus isolates with dual resistance mutations (NA-H275Y and PA-I38T) was similar to wild type virus (WT) in vitro, but reduced in the upper respiratory tracts of challenged ferrets. The dual-mutant virus transmitted well between ferrets in an airborne transmission model, but was outcompeted by the WT when the two viruses were co-administered. These results indicate the dual-mutant virus had a moderate loss of viral fitness compared to the WT virus, suggesting that while person-to-person transmission of the dual-resistant virus may be possible, widespread community transmission is unlikely., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
16. Toll-Like Receptor 7 Agonist RG7854 Mediates Therapeutic Efficacy and Seroconversion in Woodchucks With Chronic Hepatitis B.
- Author
-
Wildum S, Korolowicz KE, Suresh M, Steiner G, Dai L, Li B, Yon C, De Vera Mudry MC, Regenass-Lechner F, Huang X, Hong X, Murreddu MG, Kallakury BV, Young JAT, and Menne S
- Subjects
- Animals, Antigens, Viral, Antiviral Agents therapeutic use, Hepatitis B, Chronic immunology, Hepatitis B, Chronic veterinary, Marmota, Seroconversion, Toll-Like Receptor 7 agonists
- Abstract
Conventional treatment of chronic hepatitis B (CHB) is rarely curative due to the immunotolerant status of patients. RG7854 is an oral double prodrug of a toll-like receptor 7 (TLR7) agonist that is developed for the treatment of CHB. The therapeutic efficacy, host immune response, and safety of RG7854 were evaluated in the woodchuck model of CHB. Monotreatment with the two highest RG7854 doses and combination treatment with the highest RG7854 dose and entecavir (ETV) suppressed viral replication, led to loss of viral antigens, and induced seroconversion in responder woodchucks. Since viral suppression and high-titer antibodies persisted after treatment ended, this suggested that a sustained antiviral response (SVR) was induced by RG7854 in a subset of animals. The SVR rate, however, was comparable between both treatment regimens, suggesting that the addition of ETV did not enhance the therapeutic efficacy of RG7854 although it augmented the proliferation of blood cells in response to viral antigens and magnitude of antibody titers. The induction of interferon-stimulated genes in blood by RG7854/ETV combination treatment demonstrated on-target activation of TLR7. Together with the virus-specific blood cell proliferation and the transient elevations in liver enzymes and inflammation, this suggested that cytokine-mediated non-cytolytic and T-cell mediated cytolytic mechanisms contributed to the SVR, in addition to the virus-neutralizing effects by antibody-producing plasma cells. Both RG7854 regimens were not associated with treatment-limiting adverse effects but accompanied by dose-dependent, transient neutropenia and thrombocytopenia. The study concluded that finite, oral RG7854 treatment can induce a SVR in woodchucks that is based on the retrieval of antiviral innate and adaptive immune responses. This supports future investigation of the TLR7 agonist as an immunotherapeutic approach for achieving functional cure in patients with CHB., Competing Interests: SM has received grants from F. Hoffmann-La Roche, Ltd. and serves occasionally as a paid scientific consultant to Northeastern Wildlife, Inc. GS is employed by Roche, while LD is a former employee of Roche. SW, MV, FR-L, and JY own stock in and are employed by Roche. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The authors declare that this work was funded by F. Hoffmann-La Roche, Ltd. (https://www.roche.com) who played a role in the design of the study, collection of data, analysis and interpretation of data, preparation of the manuscript, and in the decision to submit the manuscript for publication., (Copyright © 2022 Wildum, Korolowicz, Suresh, Steiner, Dai, Li, Yon, De Vera Mudry, Regenass-Lechner, Huang, Hong, Murreddu, Kallakury, Young and Menne.)
- Published
- 2022
- Full Text
- View/download PDF
17. Tocilizumab in patients hospitalised with COVID-19 pneumonia: Efficacy, safety, viral clearance, and antibody response from a randomised controlled trial (COVACTA).
- Author
-
Rosas IO, Bräu N, Waters M, Go RC, Malhotra A, Hunter BD, Bhagani S, Skiest D, Savic S, Douglas IS, Garcia-Diaz J, Aziz MS, Cooper N, Youngstein T, Sorbo LD, Zerda DJ, Ustianowski A, Gracian AC, Blyth KG, Carratalà J, François B, Benfield T, Haslem D, Bonfanti P, van der Leest CH, Rohatgi N, Wiese L, Luyt CE, Bauer RN, Cai F, Lee IT, Matharu B, Metcalf L, Wildum S, Graham E, Tsai L, and Bao M
- Abstract
Background: In COVACTA, a randomised, placebo-controlled trial in patients hospitalised with coronavirus disease-19 (COVID-19), tocilizumab did not improve 28-day mortality, but shortened hospital and intensive care unit stay. Longer-term effects of tocilizumab in patients with COVID-19 are unknown. Therefore, the efficacy and safety of tocilizumab in COVID-19 beyond day 28 and its impact on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) clearance and antibody response in COVACTA were investigated., Methods: Adults in Europe and North America hospitalised with COVID-19 ( N = 452) between April 3, 2020 and May 28, 2020 were randomly assigned (2:1) to double-blind intravenous tocilizumab or placebo and assessed for efficacy and safety through day 60. Assessments included mortality, time to hospital discharge, SARS-CoV-2 viral load in nasopharyngeal swab and serum samples, and neutralising anti-SARS-CoV-2 antibodies in serum. ClinicalTrials.gov registration: NCT04320615., Findings: By day 60, 24·5% (72/294) of patients in the tocilizumab arm and 25·0% (36/144) in the placebo arm died (weighted difference -0·5% [95% CI -9·1 to 8·0]), and 67·0% (197/294) in the tocilizumab arm and 63·9% (92/144) in the placebo arm were discharged from the hospital. Serious infections occurred in 24·1% (71/295) of patients in the tocilizumab arm and 29·4% (42/143) in the placebo arm. Median time to negative reverse transcriptase-quantitative polymerase chain reaction result in nasopharyngeal/oropharyngeal samples was 15·0 days (95% CI 14·0 to 21·0) in the tocilizumab arm and 21·0 days (95% CI 14·0 to 28·0) in the placebo arm. All tested patients had positive test results for neutralising anti-SARS-CoV-2 antibodies at day 60., Interpretation: There was no mortality benefit with tocilizumab through day 60. Tocilizumab did not impair viral clearance or host immune response, and no new safety signals were observed. Future investigations may explore potential biomarkers to optimize patient selection for tocilizumab treatment and combination therapy with other treatments., Funding: F. Hoffmann-La Roche Ltd and the US Department of Health and Human Services, Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority, under OT number HHSO100201800036C., Competing Interests: Ivan O. Rosas: Grant from Roche/Genentech during the conduct of the study; grant and personal fees from Genentech outside the submitted work; and personal fees from Boehringer and Bristol Myers Squibb outside the submitted work Norbert Bräu: Grant support to institution from Roche/Genentech during the conduct of the study; grants from Gilead Sciences outside the submitted work Ronaldo C. Go: Consulting fees from F. Hoffmann-La Roche outside the submitted work Atul Malhotra: Grants from the National Institutes of Health; personal fees from LivaNova, Corvus, and Equillium; institutional funding from RedMed outside the submitted work Bradley D. Hunter: Personal fees from Kite Pharmaceuticals and Novartis outside the submitted work Sanjay Bhagani: Grants and personal fees from Gilead Sciences, Roche, and ViiV outside the submitted work Sinisa Savic: Grants and personal fees from Novartis and SOBI outside the submitted work Ivor S. Douglas: Grant support to institution from Roche/Genentech during the conduct of the study Andrew Ustianowski: Grant support to institution from Roche/Genentech during the conduct of the study Jordi Carratalà: Grant from Roche/Genentech during the conduct of the study; grant and personal fees from Gilead Sciences outside the submitted work Thomas Benfield: Grants from Novo Nordisk Foundation, Simonsen Foundation, GlaxoSmithKline, Pfizer, Gilead, Lundbeck Foundation, Kai Hansen Foundation and Erik and Susanna Olesen's charitable fund; personal fees from GlaxoSmithKline, Pfizer, Boehringer Ingelheim, Gilead, MSD and PentaBase A/S outside the submitted work Paolo Bonfanti: Personal fees from ViiV, Gilead, Janssen, and Merck outside the submitted work. Cor H. van der Leest: Personal fees related to the submitted work; personal fees from Bristol Myers Squib, Merck Sharp & Dohme, AbbVie, Boehringer Ingelheim, Roche, and AstraZeneca outside the submitted work Charles Edouard Luyt: Grant support to institution from Roche/Genentech during the conduct of the study; personal fees from Carmat, Merck, bioMérieux, Thermo Fischer Brahms, Bayer Healthcare, and Faron outside the submitted work Rebecca N. Bauer: Employee of Genentech and holder of stock/stock options in Roche Fang Cai: Former employee of Genentech; patent pending to Genentech for biomarkers for predicting response to an IL-6 antagonist (P36367-US) Ivan T. Lee: Former clinical research fellow of Genentech/Roche; received funding from Genentech/Roche during the conduct of the study Balpreet Matharu: Employee of Roche Products Ltd Louis Metcalf: Employee of and owns shares in Roche Steffen Wildum: Employee of F Hoffmann-La Roche Emily Graham: Employee of Roche Products Ltd Larry Tsai: Employee of Genentech/Roche; unpublished patent pending for ‘Method for treating pneumonia, including COVID-19 pneumonia, with an IL-6 antagonist’ Min Bao: Grant from Biomedical Advanced Research and Development Authority (BARDA) for the COVACTA study; employee of Genentech/Roche; unpublished patent pending for ‘Method for treating pneumonia, including COVID-19 pneumonia, with an IL-6 antagonist’ All other authors have nothing to disclose., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
18. Combining baloxavir marboxil with standard-of-care neuraminidase inhibitor in patients hospitalised with severe influenza (FLAGSTONE): a randomised, parallel-group, double-blind, placebo-controlled, superiority trial.
- Author
-
Kumar D, Ison MG, Mira JP, Welte T, Hwan Ha J, Hui DS, Zhong N, Saito T, Katugampola L, Collinson N, Williams S, Wildum S, Ackrill A, Clinch B, and Lee N
- Subjects
- Antiviral Agents, Double-Blind Method, Enzyme Inhibitors therapeutic use, Humans, Morpholines, Neuraminidase therapeutic use, Pyridones, Treatment Outcome, Triazines, Dibenzothiepins therapeutic use, Influenza, Human drug therapy
- Abstract
Background: Neuraminidase inhibitors (NAIs) are considered the standard of care for hospitalised patients with influenza. We aimed to test whether combining the cap-dependent endonuclease inhibitor baloxavir marboxil (hereafter baloxavir) with standard-of-care NAIs would result in improved clinical outcomes compared with NAI monotherapy in hospitalised patients with severe influenza., Methods: We did a randomised, parallel-group, double-blind, placebo-controlled, superiority trial. Patients aged 12 years or older who were hospitalised with laboratory-confirmed influenza (by RT-PCR or a rapid test) and had a National Early Warning Score 2 (NEWS2) of 4 or greater were included. Recruitment took place in 124 centres across 25 countries. Using a permuted-block method and an interactive response system, patients were randomly assigned (2:1) to receive either baloxavir plus NAIs (hereafter the baloxavir group) or placebo plus NAIs (hereafter the control group). Participants, investigators, and those assessing outcomes were masked to group assignment. Baloxavir was administered orally on day 1 and day 4 (40 mg for bodyweight <80 kg, or 80 mg for ≥80 kg), and on day 7 if no clinical improvement had occurred by day 5. The NAIs included in this study were oseltamivir, zanamivir, and peramivir, which were selected and administered according to local standard practice. The primary endpoint was time to clinical improvement, defined as time to a NEWS2 of 2 or lower for 24 h or hospital discharge, whichever came first, based on daily assessments over the study duration of 35 days. Secondary endpoints included safety analyses. The modified intention-to-treat infected (mITTI) population (ie, all patients who were randomly assigned to treatment, received a dose of study drug, and were RT-PCR-positive for influenza at any timepoint according to the treatment assigned at randomisation) was used in all efficacy analyses. The safety population (ie, all patients who received at least one dose of study treatment, according to the treatment received) was used in the safety analyses. The trial is registered with ClinicalTrials.gov, NCT03684044., Findings: Overall, 366 patients were enrolled between Jan 8, 2019, and March 16, 2020, of whom 241 were assigned to the baloxavir group and 125 to the control group. The mITTI population comprised 322 patients, 208 in the baloxivir group and 114 in the control group. In total, 280 (87%) of these patients had influenza A infections. Median time to clinical improvement was 97·5 h (95% CI 75·9 to 117·2) in the baloxavir group and 100·2 h (75·9 to 144·4) in the control group (median difference -2·7 h [95% CI -53·4 to 25·9], p=0·467). Baloxavir plus NAI was well tolerated, and no new safety signals were observed; serious adverse events occurred in 29 (12%) of 239 patients in the baloxavir group versus 19 (15%) of 124 patients in the control group, of which one was considered related to treatment (orthostatic hypotension in a patient in the control group). Overall, four deaths (2%) occurred in the baloxavir group and seven (6%) in the control group; none were considered related to treatment., Interpretation: Combining baloxavir with NAIs did not result in superior clinical outcomes compared with NAIs alone. The combination of baloxavir plus NAI was well tolerated. The findings suggest that combination antivirals would not be routinely indicated in clinical practice for hospitalised patients with severe influenza., Funding: F Hoffmann-La Roche and the Biomedical Advanced Research and Development Authority., Competing Interests: Declaration of interests DK has received advisory fees from F Hoffmann-La Roche, Sanofi Pasteur, and GlaxoSmithKline, and a clinical trials grant from F Hoffmann-La Roche and GlaxoSmithKline. MGI received research support, paid to Northwestern University, from AiCuris, Janssen, and Shire; he is a paid consultant for Adagio, AlloVir, Celltrion, Cidara, Genentech, F Hoffmann-La Roche, Janssen, Shionogi, and Viracor Eurofins, and he is a paid member of data monitoring and safety boards for Janssen, Merck, SAB Biotherapeutics, Sequiris, Takeda, and Vitaeris. J-PM has received advisory fees from F Hoffmann-La Roche, AM-Pharma, and AKPA; lecture fees from Fresenius, MSD, Astellas, and Estor; and clinical trial grants from AKPA and Biomérieux. NL has previously received honoraria for consultancy work and speaking in educational programmes from Shionogi, Janssen, Sanofi Pasteur, Gilead Sciences Canada, F Hoffmann-La Roche, Genentech, and CIDARA Therapeutics, and travel support from Shionogi, Sanofi Pasteur, F Hoffmann-La Roche, and Genentech. LK, NC, AA, BC, and SWill are employees of Roche Products. SWild is an employee of F Hoffmann-La Roche. TW has received honoraria for consultancy work and speaking in educational programmes from AstraZeneca, Basilea, Biotest, Bayer, Boehringer Ingelheim, Berlin Chemie, GlaxoSmithKline, Infectopharm, Johnson & Johnson, MSD, Novartis, Pfizer, F Hoffmann-La Roche, and Sanofi Aventis. TS has received lecture fees from AstraZeneca, Boehringer Ingelheim, Sanofi Pasteur, Novartis, KYORIN Pharmaceutical, and Daiichi Sankyo. DSH, NZ, and JHH have nothing to declare., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
19. Evaluating the fitness of PA/I38T-substituted influenza A viruses with reduced baloxavir susceptibility in a competitive mixtures ferret model.
- Author
-
Lee LY, Zhou J, Koszalka P, Frise R, Farrukee R, Baba K, Miah S, Shishido T, Galiano M, Hashimoto T, Omoto S, Uehara T, Mifsud EJ, Collinson N, Kuhlbusch K, Clinch B, Wildum S, Barclay WS, and Hurt AC
- Subjects
- Amino Acid Substitution, Animals, Female, Ferrets, Influenza A virus drug effects, Influenza A virus isolation & purification, Male, Orthomyxoviridae Infections virology, Antiviral Agents pharmacology, Dibenzothiepins pharmacology, Disease Models, Animal, Drug Resistance, Viral, Influenza A virus genetics, Morpholines pharmacology, Orthomyxoviridae Infections drug therapy, Pyridones pharmacology, Triazines pharmacology, Virus Replication
- Abstract
Baloxavir is approved in several countries for the treatment of uncomplicated influenza in otherwise-healthy and high-risk patients. Treatment-emergent viruses with reduced susceptibility to baloxavir have been detected in clinical trials, but the likelihood of widespread occurrence depends on replication capacity and onward transmission. We evaluated the fitness of A/H3N2 and A/H1N1pdm09 viruses with the polymerase acidic (PA) I38T-variant conferring reduced susceptibility to baloxavir relative to wild-type (WT) viruses, using a competitive mixture ferret model, recombinant viruses and patient-derived virus isolates. The A/H3N2 PA/I38T virus showed a reduction in within-host fitness but comparable between-host fitness to the WT virus, while the A/H1N1pdm09 PA/I38T virus had broadly similar within-host fitness but substantially lower between-host fitness. Although PA/I38T viruses replicate and transmit between ferrets, our data suggest that viruses with this amino acid substitution have lower fitness relative to WT and this relative fitness cost was greater in A/H1N1pdm09 viruses than in A/H3N2 viruses., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: • Keiko Baba, Takashi Hashimoto, Shinya Omoto, Takao Shishido and Takeki Uehara are employees of Shionogi & Co. • Klaus Kuhlbusch, Steffen Wildum and Aeron C Hurt are employees of F. Hoffmann-La Roche. • Neil Collinson and Barry Clinch are employees of Roche Products Ltd. • Wendy Barclay has received honoraria from Roche, Sanofi Pasteur and Seqirus. • Leo YY Lee has received honoraria from Roche. • Paulina Koszalka, Jie Zhou, Rubaiyea Farrukee, Rebecca Frise, Edin Mifsud, Monica Galiano and Shahjahan Miah have nothing to disclose.
- Published
- 2021
- Full Text
- View/download PDF
20. Comprehensive assessment of amino acid substitutions in the trimeric RNA polymerase complex of influenza A virus detected in clinical trials of baloxavir marboxil.
- Author
-
Hashimoto T, Baba K, Inoue K, Okane M, Hata S, Shishido T, Naito A, Wildum S, and Omoto S
- Subjects
- Amino Acid Substitution, Antiviral Agents pharmacology, Antiviral Agents therapeutic use, DNA-Directed RNA Polymerases genetics, DNA-Directed RNA Polymerases therapeutic use, Dibenzothiepins, Drug Resistance, Viral, Humans, Influenza A Virus, H3N2 Subtype, Morpholines therapeutic use, Pyridones therapeutic use, Triazines, Influenza A Virus, H1N1 Subtype, Influenza A virus genetics, Influenza, Human drug therapy
- Abstract
Background: Baloxavir marboxil (BXM) is an approved drug that selectively targets cap-dependent endonuclease on PA subunit in the RNA polymerase complex of influenza A and B viruses. Amino acid substitutions at position 38 in the PA subunit were identified as a major pathway for reduced susceptibility to baloxavir acid (BXA), the active form of BXM. Additionally, substitutions found at positions E23, A37, and E199 in the PA subunit impact BXA susceptibility by less than 10-fold., Methods: We comprehensively evaluated the impact of novel amino acid substitutions identified in PA, PB1, and PB2 subunits in BXM clinical trials and influenza sequence databases by means of drug susceptibility and replicative capacity., Results: PA/I38N in A(H1N1)pdm09 and PA/I38R in A(H3N2) were newly identified as treatment-emergent substitutions in the CAPSTONE-2 study. The I38N substitution conferred reduced susceptibility by 24-fold, whereas replicative capacity of the I38N-substituted virus was impaired compared with the wild-type. The I38R-substituted virus was not viable in cell culture. All other mutations assessed in this extensive study did not significantly affect BXA susceptibility (< 2.4-fold change)., Conclusion: These results provide additional information on the impact of amino acid substitutions in the trimeric viral polymerase complex to BXA susceptibility and will further support influenza surveillance., (© 2020 The Authors. Influenza and Other Respiratory Viruses Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
21. Baloxavir Marboxil Single-dose Treatment in Influenza-infected Children: A Randomized, Double-blind, Active Controlled Phase 3 Safety and Efficacy Trial (miniSTONE-2).
- Author
-
Baker J, Block SL, Matharu B, Burleigh Macutkiewicz L, Wildum S, Dimonaco S, Collinson N, Clinch B, and Piedra PA
- Subjects
- Acute Disease therapy, Administration, Oral, Antiviral Agents pharmacokinetics, Child, Child, Preschool, Dibenzothiepins pharmacokinetics, Double-Blind Method, Drug Administration Schedule, Endonucleases antagonists & inhibitors, Female, Global Health, Humans, Infant, Male, Morpholines pharmacokinetics, Pyridones pharmacokinetics, Triazines pharmacokinetics, Antiviral Agents administration & dosage, Antiviral Agents therapeutic use, Dibenzothiepins administration & dosage, Dibenzothiepins therapeutic use, Influenza, Human drug therapy, Morpholines administration & dosage, Morpholines therapeutic use, Pyridones administration & dosage, Pyridones therapeutic use, Triazines administration & dosage, Triazines therapeutic use
- Abstract
Background: Baloxavir marboxil (baloxavir) is a novel, cap-dependent endonuclease inhibitor that has previously demonstrated efficacy in the treatment of influenza in adults and adolescents. We assessed the safety and efficacy of baloxavir in otherwise healthy children with acute influenza., Methods: MiniSTONE-2 (Clinicaltrials.gov: NCT03629184) was a double-blind, randomized, active controlled trial enrolling children 1-<12 years old with a clinical diagnosis of influenza. Children were randomized 2:1 to receive either a single dose of oral baloxavir or oral oseltamivir twice daily for 5 days. The primary endpoint was incidence, severity and timing of adverse events (AEs); efficacy was a secondary endpoint., Results: In total, 173 children were randomized and dosed, 115 to the baloxavir group and 58 to the oseltamivir group. Characteristics of participants were similar between treatment groups. Overall, 122 AEs were reported in 84 (48.6%) children. Incidence of AEs was similar between baloxavir and oseltamivir groups (46.1% vs. 53.4%, respectively). The most common AEs were gastrointestinal (vomiting/diarrhea) in both groups [baloxavir: 12 children (10.4%); oseltamivir: 10 children (17.2%)]. No deaths, serious AEs or hospitalizations were reported. Median time (95% confidence interval) to alleviation of signs and symptoms of influenza was similar between groups: 138.1 (116.6-163.2) hours with baloxavir versus 150.0 (115.0-165.7) hours with oseltamivir., Conclusions: Oral baloxavir is well tolerated and effective at alleviating symptoms in otherwise healthy children with acute influenza. Baloxavir provides a new therapeutic option with a simple oral dosing regimen.
- Published
- 2020
- Full Text
- View/download PDF
22. Efficacy of an Inhibitor of Hepatitis B Virus Expression in Combination With Entecavir and Interferon-α in Woodchucks Chronically Infected With Woodchuck Hepatitis Virus.
- Author
-
Menne S, Wildum S, Steiner G, Suresh M, Korolowicz K, Balarezo M, Yon C, Murreddu M, Hong X, Kallakury BV, Tucker R, Yang S, Young JAT, and Javanbakht H
- Abstract
RG7834 is a small-molecule inhibitor of hepatitis B virus (HBV) gene expression that significantly reduces the levels of hepatitis B surface antigen (HBsAg) and HBV DNA in a humanized liver HBV mouse model. In the current study, we evaluated the potency of RG7834 in the woodchuck model of chronic HBV infection, alone and in combination with entecavir (ETV) and/or woodchuck interferon-α (wIFN-α). RG7834 reduced woodchuck hepatitis virus (WHV) surface antigen (WHsAg) by a mean of 2.57 log
10 from baseline and WHV DNA by a mean of 1.71 log10 . ETV + wIFN-α reduced WHsAg and WHV DNA by means of 2.40 log10 and 6.70 log10 , respectively. The combination of RG7834, ETV, and wIFN-α profoundly reduced WHsAg and WHV DNA levels by 5.00 log10 and 7.46 log10 , respectively. However, both viral parameters rebounded to baseline after treatment was stopped and no antibody response against WHsAg was observed. Effects on viral RNAs were mainly seen with the triple combination treatment, reducing both pregenomic RNA (pgRNA) and WHsAg RNA, whereas RG7834 mainly reduced WHsAg RNA and ETV mainly affected pgRNA. When WHsAg was reduced by the triple combination, peripheral blood mononuclear cells (PBMCs) proliferated significantly in response to viral antigens, but the cellular response was diminished after WHsAg returned to baseline levels during the off-treatment period. Consistent with this, Pearson correlation revealed a strong negative correlation between WHsAg levels and PBMC proliferation in response to peptides covering the entire WHsAg and WHV nucleocapsid antigen. Conclusion: A fast and robust reduction of WHsAg by combination therapy reduced WHV-specific immune dysfunction in the periphery. However, the magnitude and/or duration of the induced cellular response were not sufficient to achieve a sustained antiviral response., (© 2020 The Authors. Hepatology Communications published by Wiley Periodicals, Inc., on behalf of the American Association for the Study of Liver Diseases.)- Published
- 2020
- Full Text
- View/download PDF
23. Baloxavir treatment of ferrets infected with influenza A(H1N1)pdm09 virus reduces onward transmission.
- Author
-
Lee LYY, Zhou J, Frise R, Goldhill DH, Koszalka P, Mifsud EJ, Baba K, Noda T, Ando Y, Sato K, Yuki AI, Shishido T, Uehara T, Wildum S, Zwanziger E, Collinson N, Kuhlbusch K, Clinch B, Hurt AC, and Barclay WS
- Subjects
- Animals, Dibenzothiepins, Female, Ferrets, Morpholines, Orthomyxoviridae Infections transmission, Orthomyxoviridae Infections virology, Pyridones, Antiviral Agents pharmacology, Influenza A Virus, H1N1 Subtype drug effects, Neuraminidase antagonists & inhibitors, Orthomyxoviridae Infections drug therapy, Oxazines pharmacology, Pyridines pharmacology, Thiepins pharmacology, Triazines pharmacology, Virus Replication drug effects, Virus Shedding drug effects
- Abstract
Influenza viruses cause seasonal outbreaks and pose a continuous pandemic threat. Although vaccines are available for influenza control, their efficacy varies each season and a vaccine for a novel pandemic virus manufactured using current technology will not be available fast enough to mitigate the effect of the first pandemic wave. Antivirals can be effective against many different influenza viruses but have not thus far been used extensively for outbreak control. Baloxavir, a recently licensed antiviral drug that targets the influenza virus endonuclease, has been shown to reduce virus shedding more effectively than oseltamivir, a widely used neuraminidase inhibitor drug. Thus it is possible that treatment with baloxavir might also interrupt onward virus transmission. To test this, we utilized the ferret model, which is the most commonly used animal model to study influenza virus transmission. We established a subcutaneous baloxavir administration method in ferrets which achieved similar pharmacokinetics to the approved human oral dose. Transmission studies were then conducted in two different locations with different experimental setups to compare the onward transmission of A(H1N1)pdm09 virus from infected ferrets treated with baloxavir, oseltamivir or placebo to naïve sentinel ferrets exposed either indirectly in adjacent cages or directly by co-housing. We found that baloxavir treatment reduced infectious viral shedding in the upper respiratory tract of ferrets compared to placebo, and reduced the frequency of transmission amongst sentinels in both experimental setups, even when treatment was delayed until 2 days post-infection. In contrast, oseltamivir treatment did not substantially affect viral shedding or transmission compared to placebo. We did not detect the emergence of baloxavir-resistant variants in treated animals or in untreated sentinels. Our results support the concept that antivirals which decrease viral shedding could also reduce influenza transmission in the community., Competing Interests: I have read the journal's policy and the authors of this manuscript have the following competing interests: Leo Yi Yang Lee, Jie Zhou, Rebecca Frise, Daniel H. Goldhill, Paulina Koszalka and Edin J. Mifsud have nothing to declare. Kaoru Baba, Takahiro Noda, Yoshinori Ando, Kenji Sato, Aoe-Ishikawa Yuki, Takao Shishido and Takeki Uehara are employees of Shionogi & Co. Ltd. Steffen Wildum, Elke Zwanziger, Neil Collinson, Klaus Kuhlbusch, Barry Clinch and Aeron C. Hurt are employees of F. Hoffmann La Roche Ltd. Wendy S. Barclay has received honoraria from Roche, Sanofi Pasteur and Seqirus.
- Published
- 2020
- Full Text
- View/download PDF
24. Machine-learning based patient classification using Hepatitis B virus full-length genome quasispecies from Asian and European cohorts.
- Author
-
Mueller-Breckenridge AJ, Garcia-Alcalde F, Wildum S, Smits SL, de Man RA, van Campenhout MJH, Brouwer WP, Niu J, Young JAT, Najera I, Zhu L, Wu D, Racek T, Hundie GB, Lin Y, Boucher CA, van de Vijver D, and Haagmans BL
- Subjects
- Carcinoma, Hepatocellular virology, Disease Progression, Female, Humans, Liver Neoplasms virology, Male, Quasispecies, DNA, Viral, Genetic Variation, Genome, Viral, Hepatitis B virus genetics, Hepatitis B, Chronic virology, Machine Learning
- Abstract
Chronic infection with Hepatitis B virus (HBV) is a major risk factor for the development of advanced liver disease including fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The relative contribution of virological factors to disease progression has not been fully defined and tools aiding the deconvolution of complex patient virus profiles is an unmet clinical need. Variable viral mutant signatures develop within individual patients due to the low-fidelity replication of the viral polymerase creating 'quasispecies' populations. Here we present the first comprehensive survey of the diversity of HBV quasispecies through ultra-deep sequencing of the complete HBV genome across two distinct European and Asian patient populations. Seroconversion to the HBV e antigen (HBeAg) represents a critical clinical waymark in infected individuals. Using a machine learning approach, a model was developed to determine the viral variants that accurately classify HBeAg status. Serial surveys of patient quasispecies populations and advanced analytics will facilitate clinical decision support for chronic HBV infection and direct therapeutic strategies through improved patient stratification.
- Published
- 2019
- Full Text
- View/download PDF
25. PAPD5/7 Are Host Factors That Are Required for Hepatitis B Virus RNA Stabilization.
- Author
-
Mueller H, Lopez A, Tropberger P, Wildum S, Schmaler J, Pedersen L, Han X, Wang Y, Ottosen S, Yang S, Young JAT, and Javanbakht H
- Subjects
- Hepatitis B drug therapy, Humans, Two-Hybrid System Techniques, Chromosomal Proteins, Non-Histone physiology, DNA-Directed DNA Polymerase physiology, Gene Expression Regulation, Viral, Hepatitis B virus physiology, Molecular Targeted Therapy, RNA Nucleotidyltransferases physiology
- Abstract
RG7834 is a potent, orally bioavailable small-molecule inhibitor of hepatitis B virus (HBV) gene expression that belongs to the dihydroquinolizinone (DHQ) chemical class and uniquely blocks production of both viral DNA and antigens. In this study, we used DHQ compounds as tools in a compound-based adaptation version of the yeast three-hybrid screen to identify the cognate cellular protein targets, the non-canonical poly(A) RNA polymerase associated domain containing proteins 5 and 7 (PAPD5 and PAPD7). Interaction with RG7834 was mapped to the catalytic domains of the two cellular enzymes. The role of PAPD5 and PAPD7 in HBV replication was confirmed by oligonucleotide-mediated knockdown studies that phenocopied the result seen with RG7834-treated HBV-infected hepatocytes. The greatest effect on HBV gene expression was seen when PAPD5 and PAPD7 mRNAs were simultaneously knocked down, suggesting that the two cellular proteins play a redundant role in maintaining HBV mRNA levels. In addition, as seen previously with RG7834 treatment, PAPD5 and PAPD7 knockdown led to destabilization and degradation of HBV mRNA without impacting production of viral RNA transcripts. Conclusion: We identify PAPD5 and PAPD7 as cellular host factors required for HBV RNA stabilization and as therapeutic targets for the HBV cure., (© 2018 by the American Association for the Study of Liver Diseases.)
- Published
- 2019
- Full Text
- View/download PDF
26. A novel orally available small molecule that inhibits hepatitis B virus expression.
- Author
-
Mueller H, Wildum S, Luangsay S, Walther J, Lopez A, Tropberger P, Ottaviani G, Lu W, Parrott NJ, Zhang JD, Schmucki R, Racek T, Hoflack JC, Kueng E, Point F, Zhou X, Steiner G, Lütgehetmann M, Rapp G, Volz T, Dandri M, Yang S, Young JAT, and Javanbakht H
- Subjects
- Administration, Oral, Animals, Biological Availability, DNA, Viral isolation & purification, Disease Models, Animal, Mice, Treatment Outcome, Viral Load drug effects, Antiviral Agents administration & dosage, Antiviral Agents adverse effects, Antiviral Agents pharmacokinetics, Gene Expression Regulation, Viral drug effects, Hepatitis B virus drug effects, Hepatitis B virus genetics, Hepatitis B, Chronic drug therapy, Hepatitis B, Chronic virology, Small Molecule Libraries administration & dosage, Small Molecule Libraries adverse effects, Small Molecule Libraries pharmacokinetics
- Abstract
Background & Aims: The hallmarks of chronic HBV infection are a high viral load (HBV DNA) and even higher levels (>100-fold in excess of virions) of non-infectious membranous particles containing the tolerogenic viral S antigen (HBsAg). Currently, standard treatment effectively reduces viremia but only rarely results in a functional cure (defined as sustained HBsAg loss). There is an urgent need to identify novel therapies that reduce HBsAg levels and restore virus-specific immune responsiveness in patients. We report the discovery of a novel, potent and orally bioavailable small molecule inhibitor of HBV gene expression (RG7834)., Methods: RG7834 antiviral characteristics and selectivity against HBV were evaluated in HBV natural infection assays and in a urokinase-type plasminogen activator/severe combined immunodeficiency humanized mouse model of HBV infection, either alone or in combination with entecavir., Results: Unlike nucleos(t)ide therapies, which reduce viremia but do not lead to an effective reduction in HBV antigen expression, RG7834 significantly reduced the levels of viral proteins (including HBsAg), as well as lowering viremia. Consistent with its proposed mechanism of action, time course RNA-seq analysis revealed a fast and selective reduction in HBV mRNAs in response to RG7834 treatment. Furthermore, oral treatment of HBV-infected humanized mice with RG7834 led to a mean HBsAg reduction of 1.09 log
10 compared to entecavir, which had no significant effect on HBsAg levels. Combination of RG7834, entecavir and pegylated interferon α-2a led to significant reductions of both HBV DNA and HBsAg levels in humanized mice., Conclusion: We have identified a novel oral HBV viral gene expression inhibitor that blocks viral antigen and virion production, that is highly selective for HBV, and has a unique antiviral profile that is clearly differentiated from nucleos(t)ide analogues., Lay Summary: We discovered a novel small molecule viral expression inhibitor that is highly selective for HBV and unlike current therapy inhibits the expression of viral proteins by specifically reducing HBV mRNAs. RG7834 can therefore potentially provide anti-HBV benefits and increase HBV cure rates, by direct reduction of viral agents needed to complete the viral life cycle, as well as a reduction of viral agents involved in evasion of the host immune responses., (Copyright © 2017 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
27. Heteroaryldihydropyrimidine (HAP) and Sulfamoylbenzamide (SBA) Inhibit Hepatitis B Virus Replication by Different Molecular Mechanisms.
- Author
-
Zhou Z, Hu T, Zhou X, Wildum S, Garcia-Alcalde F, Xu Z, Wu D, Mao Y, Tian X, Zhou Y, Shen F, Zhang Z, Tang G, Najera I, Yang G, Shen HC, Young JA, and Qin N
- Subjects
- Antiviral Agents pharmacology, Benzamides chemistry, Binding Sites, Capsid metabolism, Crystallography, X-Ray, DNA Replication drug effects, Hep G2 Cells, Hepatitis B virus drug effects, Humans, Ligands, Mutation genetics, Photoaffinity Labels, Pyrimidines chemistry, Viral Proteins chemistry, Benzamides pharmacology, Hepatitis B virus physiology, Pyrimidines pharmacology, Virus Replication drug effects
- Abstract
Heteroaryldihydropyrimidine (HAP) and sulfamoylbenzamide (SBA) are promising non-nucleos(t)ide HBV replication inhibitors. HAPs are known to promote core protein mis-assembly, but the molecular mechanism of abnormal assembly is still elusive. Likewise, the assembly status of core protein induced by SBA remains unknown. Here we show that SBA, unlike HAP, does not promote core protein mis-assembly. Interestingly, two reference compounds HAP_R01 and SBA_R01 bind to the same pocket at the dimer-dimer interface in the crystal structures of core protein Y132A hexamer. The striking difference lies in a unique hydrophobic subpocket that is occupied by the thiazole group of HAP_R01, but is unperturbed by SBA_R01. Photoaffinity labeling confirms the HAP_R01 binding pose at the dimer-dimer interface on capsid and suggests a new mechanism of HAP-induced mis-assembly. Based on the common features in crystal structures we predict that T33 mutations generate similar susceptibility changes to both compounds. In contrast, mutations at positions in close contact with HAP-specific groups (P25A, P25S, or V124F) only reduce susceptibility to HAP_R01, but not to SBA_R01. Thus, HAP and SBA are likely to have distinctive resistance profiles. Notably, P25S and V124F substitutions exist in low-abundance quasispecies in treatment-naïve patients, suggesting potential clinical relevance.
- Published
- 2017
- Full Text
- View/download PDF
28. Differential regulation of NF-κB-mediated proviral and antiviral host gene expression by primate lentiviral Nef and Vpu proteins.
- Author
-
Sauter D, Hotter D, Van Driessche B, Stürzel CM, Kluge SF, Wildum S, Yu H, Baumann B, Wirth T, Plantier JC, Leoz M, Hahn BH, Van Lint C, and Kirchhoff F
- Subjects
- Animals, Gene Expression, HIV-1 genetics, HIV-1 metabolism, I-kappa B Kinase metabolism, I-kappa B Proteins metabolism, Immunity, Innate physiology, Lentiviruses, Primate genetics, Viral Proteins genetics, Lentiviruses, Primate metabolism, NF-kappa B metabolism, Viral Proteins metabolism
- Abstract
NF-κB is essential for effective transcription of primate lentiviral genomes and also activates antiviral host genes. Here, we show that the early protein Nef of most primate lentiviruses enhances NF-κB activation. In contrast, the late protein Vpu of HIV-1 and its simian precursors inhibits activation of NF-κB, even in the presence of Nef. Although this effect of Vpu did not correlate with its ability to interact with β-TrCP, it involved the stabilization of IκB and reduced nuclear translocation of p65. Interestingly, however, Vpu did not affect casein kinase II-mediated phosphorylation of p65. Lack of Vpu was associated with increased NF-κB activation and induction of interferon and interferon-stimulated genes (ISGs) in HIV-1-infected T cells. Thus, HIV-1 and its simian precursors employ Nef to boost NF-κB activation early during the viral life cycle to initiate proviral transcription, while Vpu is used to downmodulate NF-κB-dependent expression of ISGs at later stages., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
29. In vitro drug combination studies of Letermovir (AIC246, MK-8228) with approved anti-human cytomegalovirus (HCMV) and anti-HIV compounds in inhibition of HCMV and HIV replication.
- Author
-
Wildum S, Zimmermann H, and Lischka P
- Subjects
- Cell Line, Drug Combinations, Drug Resistance, Viral, Humans, Virus Replication drug effects, Acetates pharmacology, Anti-HIV Agents pharmacology, Antiviral Agents pharmacology, Cytomegalovirus drug effects, Quinazolines pharmacology
- Abstract
Despite modern prevention and treatment strategies, human cytomegalovirus (HCMV) remains a common opportunistic pathogen associated with serious morbidity and mortality in immunocompromised individuals, such as transplant recipients and AIDS patients. All drugs currently licensed for the treatment of HCMV infection target the viral DNA polymerase and are associated with severe toxicity issues and the emergence of drug resistance. Letermovir (AIC246, MK-8228) is a new anti-HCMV agent in clinical development that acts via a novel mode of action and has demonstrated anti-HCMV activity in vitro and in vivo. For the future, drug combination therapies, including letermovir, might be indicated under special medical conditions, such as the emergence of multidrug-resistant virus strains in transplant recipients or in HCMV-HIV-coinfected patients. Accordingly, knowledge of the compatibility of letermovir with other HCMV or HIV antivirals is of medical importance. Here, we evaluated the inhibition of HCMV replication by letermovir in combination with all currently approved HCMV antivirals using cell culture checkerboard assays. In addition, the effects of letermovir on the antiviral activities of selected HIV drugs, and vice versa, were analyzed. Using two different mathematical techniques to analyze the experimental data, (i) additive effects were observed for the combination of letermovir with anti-HCMV drugs and (ii) no interaction was found between letermovir and anti-HIV drugs. Since none of the tested drug combinations significantly antagonized letermovir efficacy (or vice versa), our findings suggest that letermovir may offer the potential for combination therapy with the tested HCMV and HIV drugs., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
30. In vitro and in vivo activities of AIC292, a novel HIV-1 nonnucleoside reverse transcriptase inhibitor.
- Author
-
Wildum S, Paulsen D, Thede K, Ruebsamen-Schaeff H, and Zimmermann H
- Subjects
- Animals, Anti-HIV Agents chemical synthesis, Cell Line, Clinical Trials, Phase I as Topic, Drug Resistance, Viral drug effects, Drug Synergism, Drug Therapy, Combination, HIV Infections virology, HIV Reverse Transcriptase genetics, HIV Reverse Transcriptase metabolism, HIV-1 growth & development, Heterografts drug effects, Heterografts virology, Humans, Imidazolidines chemical synthesis, Mice, Mutation, Pyrazoles chemical synthesis, Reverse Transcriptase Inhibitors chemical synthesis, Anti-HIV Agents pharmacology, HIV Infections drug therapy, HIV Reverse Transcriptase antagonists & inhibitors, HIV-1 drug effects, Imidazolidines pharmacology, Pyrazoles pharmacology, Reverse Transcriptase Inhibitors pharmacology
- Abstract
Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are important and frequently used elements of highly active antiretroviral therapy (HAART) for the treatment of human immunodeficiency virus type 1 (HIV-1) infection. However, the development of drug resistance, as well as the side effects of existing drugs, defines a medical need for novel NNRTIs with excellent tolerability, improved activity against NNRTI-resistant viruses, and a low barrier to resistance. Within the chemical class of diarylpyrazole-[imidazolidinone]-carboxamides, AIC292 was identified as a promising novel HIV-1 NNRTI and has successfully completed single-dose clinical phase I studies. Here, we report on the antiviral activity of AIC292, evaluated in vitro against wild-type and NNRTI-resistant HIV-1 isolates and in vivo using an engineered mouse xenograft model. AIC292 inhibited wild-type HIV-1 laboratory strains at low nanomolar concentrations, was well tolerated in different cell lines, and showed excellent selectivity in a lead profiling screen. In addition, activity of AIC292 could be demonstrated against a broad panel of wild-type HIV-1 group M and group O clinical isolates. AIC292 also retained activity against viruses harboring NNRTI resistance-associated mutations (RAMs), including the most prevalent variants, K103N, Y181C, and G190A. Interestingly, viruses bearing the L100I RAM were hypersusceptible to AIC292. Two-drug combination assays showed no antagonistic interactions between AIC292 and representative marketed HIV drugs with regard to antiviral activity. Furthermore, AIC292 displayed potent antiviral in vivo efficacy in a mouse xenograft model when applied once daily. Taken together, these data show that AIC292 represents a molecule with the antiviral properties of a novel NNRTI for the treatment of HIV-1 infection.
- Published
- 2013
- Full Text
- View/download PDF
31. In vitro evaluation of the activities of the novel anticytomegalovirus compound AIC246 (letermovir) against herpesviruses and other human pathogenic viruses.
- Author
-
Marschall M, Stamminger T, Urban A, Wildum S, Ruebsamen-Schaeff H, Zimmermann H, and Lischka P
- Subjects
- Antiviral Agents chemistry, Cytomegalovirus isolation & purification, Drug Resistance, Viral drug effects, Herpesviridae classification, Herpesviridae drug effects, Humans, Microbial Sensitivity Tests, Quinazolines chemistry, Species Specificity, Virus Diseases virology, Viruses classification, Viruses drug effects, Antiviral Agents pharmacology, Cytomegalovirus drug effects, Cytomegalovirus Infections virology, Quinazolines pharmacology
- Abstract
AIC246 (letermovir) is a potent anticytomegalovirus drug in clinical development. Here, we report a consistent antiviral efficacy of AIC246 against human cytomegalovirus laboratory strains, clinical isolates, and virus variants resistant to approved drugs. Furthermore, we describe a remarkable selectivity of AIC246 for human cytomegaloviruses compared to that of other alpha-, beta-, or gammaherpesviruses or nonrelated pathogenic viruses, including adeno-, hepadna-, retro-, orthomyxo-, and flaviviruses. Our data confirm and support an excellent and selective anticytomegaloviral activity of AIC246.
- Published
- 2012
- Full Text
- View/download PDF
32. Semen-derived amyloid fibrils drastically enhance HIV infection.
- Author
-
Münch J, Rücker E, Ständker L, Adermann K, Goffinet C, Schindler M, Wildum S, Chinnadurai R, Rajan D, Specht A, Giménez-Gallego G, Sánchez PC, Fowler DM, Koulov A, Kelly JW, Mothes W, Grivel JC, Margolis L, Keppler OT, Forssmann WG, and Kirchhoff F
- Subjects
- Acid Phosphatase, Amyloid isolation & purification, Animals, Animals, Genetically Modified, Humans, Peptide Library, Rats, Semen enzymology, Semen physiology, Viral Load, Amyloid physiology, HIV Infections transmission, Peptide Fragments physiology, Protein Tyrosine Phosphatases chemistry, Protein Tyrosine Phosphatases physiology, Semen metabolism, Sexually Transmitted Diseases, Viral metabolism
- Abstract
Sexual intercourse is the major route of HIV transmission. To identify endogenous factors that affect the efficiency of sexual viral transmission, we screened a complex peptide/protein library derived from human semen. We show that naturally occurring fragments of the abundant semen marker prostatic acidic phosphatase (PAP) form amyloid fibrils. These fibrils, termed Semen-derived Enhancer of Virus Infection (SEVI), capture HIV virions and promote their attachment to target cells, thereby enhancing the infectious virus titer by several orders of magnitude. Physiological concentrations of SEVI amplified HIV infection of T cells, macrophages, ex vivo human tonsillar tissues, and transgenic rats in vivo, as well as trans-HIV infection of T cells by dendritic or epithelial cells. Amyloidogenic PAP fragments are abundant in seminal fluid and boost semen-mediated enhancement of HIV infection. Thus, they may play an important role in sexual transmission of HIV and could represent new targets for its prevention.
- Published
- 2007
- Full Text
- View/download PDF
33. Two DEAD-box proteins may be part of RNA-dependent high-molecular-mass protein complexes in Arabidopsis mitochondria.
- Author
-
Matthes A, Schmidt-Gattung S, Köhler D, Forner J, Wildum S, Raabe M, Urlaub H, and Binder S
- Subjects
- Arabidopsis genetics, Arabidopsis metabolism, Cells, Cultured, Cold Temperature, DEAD-box RNA Helicases genetics, Gene Expression Regulation, Plant, Mitochondria metabolism, Promoter Regions, Genetic, Transcription, Genetic, Arabidopsis enzymology, DEAD-box RNA Helicases metabolism, Mitochondria enzymology, Multiprotein Complexes metabolism, RNA metabolism
- Abstract
Posttranscriptional processes are important for regulation of gene expression in plant mitochondria. DEAD-box proteins, which form a huge protein family with members from all kingdoms, are fundamental components in virtually all types of processes in RNA metabolism. Two members of this protein family, designated PMH1 and PMH2 (for PUTATIVE MITOCHONDRIAL RNA HELICASE), were analyzed and characterized in mitochondria of Arabidopsis (Arabidopsis thaliana). Green fluorescent protein tagging with N-terminal PMH1 and PMH2 sequences supports the mitochondrial localization of these proteins. Northern experiments, as well as histochemical beta-glucuronidase staining of transgenic plants carrying respective promoter:beta-glucuronidase fusion constructs, revealed differing transcription patterns for the two genes. In response to cold, however, transcript levels of both genes increased. Immunodetection analyses of mitochondrial protein complexes after two-dimensional blue native/urea SDS-PAGE and after fractionation on sucrose gradients strongly suggest that one or both proteins are part of RNA-dependent complexes. Cold treatment of cell cultures or solubilization of mitochondria in the presence of MgCl(2) favored the detection of high-molecular-mass complexes. This study paves the way for detailed analysis of high-molecular-mass complexes in mitochondria of higher plants.
- Published
- 2007
- Full Text
- View/download PDF
34. Nef alleles from children with non-progressive HIV-1 infection modulate MHC-II expression more efficiently than those from rapid progressors.
- Author
-
Schindler M, Wildum S, Casartelli N, Doria M, and Kirchhoff F
- Subjects
- Acquired Immunodeficiency Syndrome genetics, Acquired Immunodeficiency Syndrome immunology, Antigens, Differentiation, B-Lymphocyte genetics, Antigens, Viral genetics, Antigens, Viral immunology, CD4 Antigens genetics, CD4-Positive T-Lymphocytes immunology, Down-Regulation genetics, Gene Expression Regulation, Viral genetics, HIV Infections immunology, HIV Long-Term Survivors, HeLa Cells, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I immunology, Histocompatibility Antigens Class II immunology, Humans, Infant, Newborn, Jurkat Cells, Lymphocyte Activation genetics, Up-Regulation genetics, Genes, nef genetics, HIV Infections genetics, HIV-1 genetics, Histocompatibility Antigens Class II genetics
- Abstract
Background: It has been established that defective nef genes and differences in the Nef-mediated downmodulation of CD4 and MHC-I cell surface expression can be associated with different rates of HIV-1 disease progression., Objective: To evaluate whether nef alleles derived from perinatally HIV-1-infected children showing no, slow or rapid disease progression differ in their abilities to downmodulate mature MHC-II or to upregulate the invariant chain (Ii) associated with immature MHC-II complexes., Methods: Nef alleles derived from HIV-1-infected children were cloned into expression vectors and proviral HIV-1 constructs co-expressing Nef and enhanced green fluorescence protein via an internal ribosomal entry site. Nef-mediated modulation of CD4, MHC-I, MHC-II or Ii surface expression was analysed by flow cytometric analysis of Jurkat T cells, monocytic THP-1 cells, CD4 T cells and macrophages transduced with vesicular stomatitis virus G-pseudotyped HIV-1 nef variants or transiently transfected HeLa class II transactivator cells., Results: : Nef alleles derived from HIV-1-infected children with non-progressive infection were significantly more active in the upregulation of Ii and downregulation of MHC-II than those derived from rapid progressors., Conclusion: Nef alleles particularly active in interfering with MHC-II antigen presentation are more frequently found in perinatally HIV-1-infected non-progressors than rapid progressors. Possibly in the context of an immature host immune system, strongly impaired MHC-II function might contribute to lower levels of immune activation and a decelerated loss of CD4 T cells.
- Published
- 2007
- Full Text
- View/download PDF
35. Primary human immunodeficiency virus type 1 nef alleles show major differences in pathogenicity in transgenic mice.
- Author
-
Priceputu E, Hanna Z, Hu C, Simard MC, Vincent P, Wildum S, Schindler M, Kirchhoff F, and Jolicoeur P
- Subjects
- Amino Acid Sequence, Animals, Apoptosis genetics, Base Sequence, CD4 Antigens genetics, CD4 Antigens metabolism, CD4-Positive T-Lymphocytes immunology, Cell Line, DNA Primers, Flow Cytometry, HIV-1 genetics, Humans, Immunologic Memory genetics, Immunoprecipitation, Lymphocyte Activation, Mice, Mice, Transgenic, Molecular Sequence Data, Regulatory Elements, Transcriptional genetics, Sequence Analysis, DNA, nef Gene Products, Human Immunodeficiency Virus, Alleles, Gene Expression Regulation, Viral, Gene Products, nef genetics, HIV Infections genetics, HIV-1 pathogenicity
- Abstract
We previously reported that the human immunodeficiency virus type 1 NL4-3 Nef is necessary and sufficient to induce a severe AIDS-like disease in transgenic (Tg) mice when the protein is expressed under the regulatory sequences of the human CD4 gene. We have now assayed additional Nef alleles (SF2, JR-CSF, YU10x, and NL4-3 [T71R] Nef alleles), including some from long-term nonprogressors (AD-93, 032an, and 039nm alleles) in the same Tg system and compared their pathogenicities. All these Nef alleles downregulated cell surface CD4 in human cells in vitro and also, with the exception of Nef(YU10x), in Tg CD4(+) T cells. Depletion of double-positive and single-positive thymocytes occurred with all alleles but was less pronounced in Nef(YU10x) Tg mice. A loss of peripheral CD4(+) T cells was observed with all alleles but was minimal in Nef(YU10x) Tg mice. In Nef(032an) and Nef(SF2) Tg mice, T-cell loss was severe despite lower levels of Tg expression, suggesting a higher virulence of these alleles. All Nef alleles except the Nef(YU10x) and Nef(NL4-3(T71R)) alleles induced an enhanced activated memory (CD25(+) CD69(+) CD44(high) CD45RB(low) CD62L(low)) and apoptotic phenotype. Also, all could interact with and/or activate PAK2 except the Nef(JR-CSF) allele. Organ (lung and kidney) diseases were present in Nef(NL4-3(T71R)), Nef(032an), Nef(039nm), and Nef(SF2) Tg mice, despite very low levels of Tg expression for the last strain. However, no organ disease or minimal organ disease developed in Nef(YU10x) and Nef(AD-93) Tg mice and Nef(JR-CSF) Tg mice, respectively, despite high levels of Tg expression. Our data show that important differences in the pathogenicities of various Nef alleles can be scored in Tg mice. Interestingly, our results also revealed that some phenotypes can segregate independently, such as CD4(+) T-cell depletion and activation, as well as severe depletion of thymic CD4(+) T cells and peripheral CD4(+) T cells. Therefore, expression of Nef alleles in Tg mice under the CD4C regulatory elements represents a novel assay for measuring their pathogenicity. Because of the very high similarity of this murine AIDS-like disease to human AIDS, this assay may have a predictive value regarding the behavior of Nef in infected humans.
- Published
- 2007
- Full Text
- View/download PDF
36. Mapping of mitochondrial mRNA termini in Arabidopsis thaliana: t-elements contribute to 5' and 3' end formation.
- Author
-
Forner J, Weber B, Thuss S, Wildum S, and Binder S
- Subjects
- Arabidopsis enzymology, Base Sequence, Electron Transport Complex IV genetics, Endoribonucleases metabolism, Gene Expression Regulation, Plant, Mitochondria enzymology, Molecular Sequence Data, Nucleic Acid Conformation, RNA metabolism, RNA 3' End Processing, RNA, Messenger metabolism, RNA, Mitochondrial, RNA, Plant metabolism, RNA, Transfer chemistry, Reverse Transcriptase Polymerase Chain Reaction, Arabidopsis genetics, RNA chemistry, RNA Processing, Post-Transcriptional, RNA, Messenger chemistry, RNA, Plant chemistry
- Abstract
With CR-RT-PCR as primary approach we mapped the 5' and 3' transcript ends of all mitochondrial protein-coding genes in Arabidopsis thaliana. Almost all transcripts analyzed have single major 3' termini, while multiple 5' ends were found for several genes. Some of the identified 5' ends map within promoter motifs suggesting these ends to be derived from transcription initiation while the majority of the 5' termini seems to be generated post-transcriptionally. Assignment of the extremities of 5' leader RNAs revealed clear evidence for an endonucleolytic generation of the major cox1 and atp9 5' mRNA ends. tRNA-like structures, so-called t-elements, are associated either with 5' or with 3' termini of several mRNAs. These secondary structures most likely act as cis-signals for endonucleolytic cleavages by RNase Z and/or RNase P. Since no conserved sequence motif is evident at post-transcriptionally derived ends, we suggest t-elements, stem-loops and probably complex higher order structures as cis-elements for processing. This analysis provides novel insights into 5' and 3' end formation of mRNAs. In addition, the complete transcript map is a substantial and important basis for future studies of gene expression in mitochondria of higher plants.
- Published
- 2007
- Full Text
- View/download PDF
37. Contribution of Vpu, Env, and Nef to CD4 down-modulation and resistance of human immunodeficiency virus type 1-infected T cells to superinfection.
- Author
-
Wildum S, Schindler M, Münch J, and Kirchhoff F
- Subjects
- Apoptosis, CD4 Antigens analysis, CD4-Positive T-Lymphocytes chemistry, CD4-Positive T-Lymphocytes immunology, Down-Regulation, Gene Products, env genetics, Gene Products, env physiology, Gene Products, nef genetics, Gene Products, nef physiology, HIV Infections immunology, Human Immunodeficiency Virus Proteins, Humans, Jurkat Cells, Superinfection immunology, Viral Proteins genetics, Viral Regulatory and Accessory Proteins genetics, Viral Regulatory and Accessory Proteins physiology, nef Gene Products, Human Immunodeficiency Virus, CD4 Antigens metabolism, CD4-Positive T-Lymphocytes virology, HIV Infections virology, HIV-1, Viral Proteins physiology
- Abstract
Human immunodeficiency virus type 1 (HIV-1) utilizes Vpu, Env, and Nef to down-modulate its primary CD4 receptor from the cell surface, and this function seems to be critical for the pathogenesis of AIDS. The physiological relevance of CD4 down-modulation, however, is currently not well understood. In the present study, we analyzed the kinetics of CD4 down-modulation and the susceptibility of HIV-1-infected T cells to superinfection using proviral HIV-1 constructs containing individual and combined defects in vpu, env, and nef and expressing red or green fluorescent proteins. T cells infected with HIV-1 mutants containing functional nef genes expressed low surface levels of CD4 from the first moment that viral gene expression became detectable. In comparison, Vpu and Env had only minor to moderate effects on CD4 during later stages of infection. Consistent with these quantitative differences, Nef inhibited superinfection more efficiently than Vpu and Env. Notably, nef alleles from AIDS patients were more effective in preventing superinfection than those derived from a nonprogressor of HIV-1 infection. Our data suggest that protection against X4-tropic HIV-1 superinfection involves both CD4-independent and CD4-dependent mechanisms of HIV-1 Nef. X4 was effectively down-regulated by simian immunodeficiency virus and HIV-2 but not by HIV-1 Nef proteins. Thus, maximal protection seems to involve an as-yet-unknown mechanism that is independent of CD4 or coreceptor down-modulation. Finally, we demonstrate that superinfected primary T cells show enhanced levels of apoptosis. Accordingly, one reason that HIV-1 inhibits CD4 surface expression and superinfection is to prevent premature cell death in order to expand the period of effective virus production.
- Published
- 2006
- Full Text
- View/download PDF
38. Importance of the N-distal AP-2 binding element in Nef for simian immunodeficiency virus replication and pathogenicity in rhesus macaques.
- Author
-
Brenner M, Münch J, Schindler M, Wildum S, Stolte N, Stahl-Hennig C, Fuchs D, Mätz-Rensing K, Franz M, Heeney J, Ten Haaft P, Swigut T, Hrecka K, Skowronski J, and Kirchhoff F
- Subjects
- Acute Disease, Alleles, Amino Acid Sequence, Animals, Binding Sites, Cell Line, Gene Products, nef genetics, Genetic Variation genetics, Humans, Lentivirus Infections genetics, Lentivirus Infections metabolism, Lentivirus Infections virology, Macaca mulatta genetics, Molecular Sequence Data, Protein Binding, Sequence Alignment, Time Factors, Adaptor Protein Complex 2 metabolism, Gene Products, nef chemistry, Gene Products, nef metabolism, Macaca mulatta virology, Simian Immunodeficiency Virus pathogenicity, Simian Immunodeficiency Virus physiology, Virus Replication
- Abstract
Point mutations in SIVmac239 Nef disrupting CD4 downmodulation and enhancement of virion infectivity attenuate viral replication in acutely infected rhesus macaques, but changes selected later in infection fully restore Nef function (A. J. Iafrate et al., J. Virol. 74:9836-9844, 2000). To further evaluate the relevance of these Nef functions for viral persistence and disease progression, we analyzed an SIVmac239 Nef mutant containing a deletion of amino acids Q64 to N67 (delta64-67Nef). This mutation inactivates the N-distal AP-2 clathrin adaptor binding element and disrupts the abilities of Nef to downregulate CD4, CD28 and CXCR4 and to stimulate viral replication in vitro. However, it does not impair the downmodulation of CD3 and class I major histocompatibility complex (MHC-I) or MHC-II and the upregulation of the MHC-II-associated invariant chain, and it has only a moderate effect on the enhancement of virion infectivity. Replication of the delta64-67Nef variant in acutely infected macaques was intermediate between grossly nef-deleted and wild-type SIVmac239. Subsequently, three of six macaques developed moderate to high viral loads and developed disease, whereas the remaining animals efficiently controlled SIV replication and showed a more attenuated clinical course of infection. Sequence analysis revealed that the deletion in nef was not repaired in any of these animals. However, some changes that slightly enhanced the ability of Nef to downmodulate CD4 and moderately increased Nef-mediated enhancement of viral replication and infectivity in vitro were observed in macaques developing high viral loads. Our results imply that both the Nef functions that were disrupted by the delta64-67 mutation and the activities that remained intact contribute to viral pathogenicity.
- Published
- 2006
- Full Text
- View/download PDF
39. Effect of R77Q, R77A and R80A changes in Vpr on HIV-1 replication and CD4 T cell depletion in human lymphoid tissue ex vivo.
- Author
-
Rajan D, Wildum S, Rücker E, Schindler M, and Kirchhoff F
- Subjects
- Apoptosis, Blotting, Western, CD4 Lymphocyte Count, HIV Infections immunology, HIV Infections pathology, HIV-1 immunology, HIV-1 pathogenicity, HIV-1 physiology, Humans, Lymphoid Tissue immunology, Lymphoid Tissue pathology, Polymerase Chain Reaction methods, Tissue Culture Techniques, Virus Replication, CD4-Positive T-Lymphocytes immunology, Genes, vpr genetics, HIV Infections virology, HIV-1 genetics, Lymphoid Tissue virology, Point Mutation
- Abstract
Background: It has been suggested that mutations of R77A and R80A in the HIV-1 viral protein R (Vpr) impair its proapoptotic activity and that a naturally occurring R77Q variation is associated with non-progressive HIV-1 infection., Rationale: To assess the effect of Vpr R77Q, R77A and R80A mutations on the efficiency of CCR5(R5)- and CXCR4(X4)-tropic HIV-1 replication and cytopathicity in human lymphoid tissue (HLT)., Methods: Vpr mutants of the X4-tropic HIV-1 NL4-3 clone and an R5-tropic derivative were generated by PCR mutagenesis. Virus stocks established by transfection of 293T cells were used to infect macrophages and ex vivo HLT. HIV-1 replication was assessed by measuring p24 core antigen in the culture supernatants and CD4 T-cell depletion and apoptosis were measured by flow cytometric analysis., Results: The R5-tropic HIV-1 Vpr mutants replicated with slightly (R77A, R77Q) to moderately (R80A) reduced efficiency in ex vivo-infected HLT and macrophages. In comparison, the changes in Vpr had negligible effects on replication of the X4-tropic forms in lymphatic tissues. Mutation of R77Q and R80A reduced apoptosis of HIV-1-infected cells in ex vivo-infected HLT independently of the viral coreceptor tropism. However, only the R5-tropic HIV-1 Vpr mutants caused markedly less CD4 T-cell depletion than wild-type HIV-1 at the end of ex vivo HLT culture., Conclusions: The observation that Vpr R77Q reduces the cytopathicity of R5-tropic HIV-1 in lymphoid tissues supports a role in non-progressive HIV-1 infection but the attenuating effects might be dependent on the viral subtype and coreceptor tropism.
- Published
- 2006
- Full Text
- View/download PDF
40. The role of upstream U3 sequences in HIV-1 replication and CD4+ T cell depletion in human lymphoid tissue ex vivo.
- Author
-
Münch J, Rajan D, Rücker E, Wildum S, Adam N, and Kirchhoff F
- Subjects
- CD4 Lymphocyte Count, Cell Line, Cells, Cultured, Gene Products, nef analysis, Genes, nef, HIV Core Protein p24 analysis, HIV Long Terminal Repeat physiology, HIV Reverse Transcriptase analysis, Humans, Mutation, Regulatory Elements, Transcriptional, nef Gene Products, Human Immunodeficiency Virus, CD4-Positive T-Lymphocytes immunology, HIV Long Terminal Repeat genetics, HIV-1 genetics, HIV-1 physiology, Palatine Tonsil virology, Virus Replication
- Abstract
The LTRs of all primate lentiviruses contain long U3 regions overlapping the nef gene. To assess the relevance of the modulatory U3 region for HIV-1 replication, we inactivated the T-rich region, the Polypurine tract and attachment (att) sequences in nef by silent mutations and inserted intact cis-regulatory elements just upstream of the core enhancer. These modifications severely truncated the U3 region and eliminated the nef overlap. The resulting HIV-1 mutants expressed functional Nef, replicated efficiently and caused CD4+ T cell depletion in ex vivo-infected lymphoid tissue suggesting that the modulatory U3 region might not be essential for efficient HIV-1 gene expression and AIDS pathogenesis.
- Published
- 2005
- Full Text
- View/download PDF
41. Primary sooty mangabey simian immunodeficiency virus and human immunodeficiency virus type 2 nef alleles modulate cell surface expression of various human receptors and enhance viral infectivity and replication.
- Author
-
Münch J, Schindler M, Wildum S, Rücker E, Bailer N, Knoop V, Novembre FJ, and Kirchhoff F
- Subjects
- Amino Acid Sequence, Animals, CD28 Antigens analysis, CD4 Antigens analysis, Cercocebus atys, HIV-2 pathogenicity, HIV-2 physiology, HeLa Cells, Histocompatibility Antigens Class I analysis, Humans, Jurkat Cells, Lymphocyte Activation, Molecular Sequence Data, Mutation, Phylogeny, Simian Immunodeficiency Virus pathogenicity, Simian Immunodeficiency Virus physiology, T-Lymphocytes immunology, Alleles, Genes, nef physiology, HIV-2 genetics, Simian Immunodeficiency Virus genetics, Virus Replication
- Abstract
The nef gene of the pathogenic simian immunodeficiency virus (SIV) mac239 clone has been well characterized. Little is known, however, about the function of nef alleles derived from naturally SIVsm-infected sooty mangabeys (Cercocebus atys) and from human immunodeficiency virus type 2 (HIV-2)-infected individuals. Addressing this, we demonstrate that, similarly to the SIVmac239 nef, primary SIVsm and HIV-2 nef alleles down-modulate cell surface expression of human CD4, CD28, CD3, and class I or II major histocompatibility complex (MHC-I or MHC-II, respectively) molecules, up-regulate surface expression of the invariant chain (Ii) associated with immature MHC-II, inhibit early T-cell activation events, and enhance virion infectivity. Both also stimulate viral replication, although HIV-2 nef alleles were less active in this assay than SIVsm nef alleles. Mutational analysis showed that a dileucine-based sorting motif in the C-proximal loop of SIV or HIV-2 Nef is critical for its effects on CD4, CD28, and Ii but dispensable for down-regulation of CD3, MHC-I, and MHC-II. The C terminus of SIV and HIV-2 Nef was exclusively required for down-modulation of MHC-I, further demonstrating that analogous functions are mediated by different domains in Nef proteins derived from different groups of primate lentiviruses. Our results demonstrate that none of the eight Nef functions investigated had been newly acquired after cross-species transmission of SIVsm from naturally infected mangabeys to humans or macaques. Notably, HIV-2 and SIVsm nef alleles efficiently down-modulate CD3 and C28 surface expression and inhibit T-cell activation more efficiently than HIV-1 nef alleles. These differences in Nef function might contribute to the relatively low levels of immune activation observed in HIV-2-infected human individuals.
- Published
- 2005
- Full Text
- View/download PDF
42. A naturally occurring variation in the proline-rich region does not attenuate human immunodeficiency virus type 1 nef function.
- Author
-
Rücker E, Münch J, Wildum S, Brenner M, Eisemann J, Margolis L, and Kirchhoff F
- Subjects
- Amino Acid Substitution, Gene Products, nef chemistry, Gene Products, nef physiology, Genes, nef, Genetic Variation, HIV-1 pathogenicity, HIV-1 physiology, HeLa Cells, Humans, Jurkat Cells, Mutagenesis, Site-Directed, Proline chemistry, Virulence genetics, Virus Replication genetics, nef Gene Products, Human Immunodeficiency Virus, Gene Products, nef genetics, HIV-1 genetics
- Abstract
We analyzed human immunodeficiency virus type 1 (HIV-1) Nef variants to further evaluate the functional relevance of the R71T substitution previously proposed to attenuate viral replication (Fackler et al., Curr. Biol. 11:1294-1299, 2001). Our results demonstrate that this variation in the proline-rich region does not significantly affect the functional activity of Nef or HIV-1 infectivity or replication., (Copyright 2004 American Society for Microbiology)
- Published
- 2004
- Full Text
- View/download PDF
43. Alterations in HIV-1 LTR promoter activity during AIDS progression.
- Author
-
Hiebenthal-Millow K, Greenough TC, Bretttler DB, Schindler M, Wildum S, Sullivan JL, and Kirchhoff F
- Subjects
- Adolescent, Adult, Base Sequence, Cell Line, Child, Child, Preschool, Disease Progression, HIV Infections virology, HIV-1 genetics, Humans, Jurkat Cells, Leukocytes, Mononuclear virology, Middle Aged, Molecular Sequence Data, U937 Cells, HIV Infections physiopathology, HIV Long Terminal Repeat genetics, HIV-1 pathogenicity, Mutation, Promoter Regions, Genetic genetics
- Abstract
HIV-1 variants evolving in AIDS patients frequently show increased replicative capacity compared to those present during early asymptomatic infection. It is known that late stage HIV-1 variants often show an expanded coreceptor tropism and altered Nef function. In the present study we investigated whether enhanced HIV-1 LTR promoter activity might also evolve during disease progression. Our results demonstrate increased LTR promoter activity after AIDS progression in 3 of 12 HIV-1-infected individuals studied. Further analysis revealed that multiple alterations in the U3 core-enhancer and in the transactivation-response (TAR) region seem to be responsible for the enhanced functional activity. Our findings show that in a subset of HIV-1-infected individuals enhanced LTR transcription contributes to the increased replicative potential of late stage virus isolates and might accelerate disease progression.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.