Extended Evaluation of Virological, Immunological and Pharmacokinetic Endpoints of CELADEN: A Randomized, Placebo-Controlled Trial of Celgosivir in Dengue Fever Patients

Cynthia Sung, Yuan Wei, Satoru Watanabe, How Sung Lee, Yok Moi Khoo, Lu Fan, Abhay P S Rathore, Kitti Wing-Ki Chan, Milly M Choy, Uma S Kamaraj, October M Sessions, Pauline Aw, Paola F de Sessions, Bernett Lee, John E Connolly, Martin L Hibberd, Dhanasekaran Vijaykrishna, Limin Wijaya, Eng Eong Ooi, Jenny Guek-Hong Low, Subhash G Vasudevan, Cynthia Sung, Yuan Wei, Satoru Watanabe, How Sung Lee, Yok Moi Khoo, Lu Fan, Abhay P S Rathore, Kitti Wing-Ki Chan, Milly M Choy, Uma S Kamaraj, October M Sessions, Pauline Aw, Paola F de Sessions, Bernett Lee, John E Connolly, Martin L Hibberd, Dhanasekaran Vijaykrishna, Limin Wijaya, Eng Eong Ooi, Jenny Guek-Hong Low, Subhash G Vasudevan

Abstract

CELADEN was a randomized placebo-controlled trial of 50 patients with confirmed dengue fever to evaluate the efficacy and safety of celgosivir (A study registered at ClinicalTrials.gov, number NCT01619969). Celgosivir was given as a 400 mg loading dose and 200 mg bid (twice a day) over 5 days. Replication competent virus was measured by plaque assay and compared to reverse transcription quantitative PCR (qPCR) of viral RNA. Pharmacokinetics (PK) correlations with viremia, immunological profiling, next generation sequence (NGS) analysis and hematological data were evaluated as exploratory endpoints here to identify possible signals of pharmacological activity. Viremia by plaque assay strongly correlated with qPCR during the first four days. Immunological profiling demonstrated a qualitative shift in T helper cell profile during the course of infection. NGS analysis did not reveal any prominent signature that could be associated with drug treatment; however the phylogenetic spread of patients' isolates underlines the importance of strain variability that may potentially confound interpretation of dengue drug trials conducted during different outbreaks and in different countries. Celgosivir rapidly converted to castanospermine (Cast) with mean peak and trough concentrations of 5727 ng/mL (30.2 μM) and 430 ng/mL (2.3 μM), respectively and cleared with a half-life of 2.5 (± 0.6) hr. Mean viral log reduction between day 2 and 4 (VLR2-4) was significantly greater in secondary dengue than primary dengue (p = 0.002). VLR2-4 did not correlate with drug AUC but showed a trend of greater response with increasing Cmin. PK modeling identified dosing regimens predicted to achieve 2.4 to 4.5 times higher Cmin. than in the CELADEN trial for only 13% to 33% increase in overall dose. A small, non-statistical trend towards better outcome on platelet nadir and difference between maximum and minimum hematocrit was observed in celgosivir-treated patients with secondary dengue infection. Optimization of the dosing regimen and patient stratification may enhance the ability of a clinical trial to demonstrate celgosivir activity in treating dengue fever based on hematological endpoints. A new clinical trial with a revised dosing regimen is slated to start in 2016 (NCT02569827). Furthermore celgosivir's potential value for treatment of other flaviruses such as Zika virus should be investigated urgently.

Trial registration: ClinicalTrials.gov NCT01619969.

Conflict of interest statement

CS, SW, APSR, KWKC, EEO, JGHL and SGV are named inventors on a patent (Novel Dosing Regimens of Celgosivir For The Treatment Of Dengue; filing date Dec 4, 2013). CS reports personal fees from 60 Degrees Pharmaceuticals for examining pharmacokinetic modelling and dosing regimen for dengue antiviral drugs. JGHL and SGV received research support from 60° Pharmaceuticals PLC and Ministry of Health of Singapore Category 1 grant for extended non-clinical studies on celgosivir.

Figures

Fig 1. Subject disposition for CELADEN study…
Fig 1. Subject disposition for CELADEN study [17].
Fig 2. Correlation between viremia by qPCR…
Fig 2. Correlation between viremia by qPCR and plaque assay.
Red filled circles–patients who received celgosivir; blue open squares–patients who received placebo; lines–linear regression to all data. Pearson correlation coefficient for all data (A)Day 1 0.79 (95% CI: 0.64 to 0.87); (B) Day 2 0.79 (95% CI: 0.65 to 0.87); (C) Day 3 0.83 (95% CI: 0.72 to 0.90); (D) Day 4 (95% CI: 0.55 to 0.83); (E) Day 5 0.36 (95% CI: 0.08 to 0.58).
Fig 3. Viremia kinetics in primary and…
Fig 3. Viremia kinetics in primary and secondary dengue patients.
Log viremia Mean (± SEM) by day and prior dengue infection status. Green solid circles connected by solid line–primary dengue; open orange squares connected by dashed line–secondary dengue. Inset: VLR 2–4 by prior dengue infection status: box 25th to 75th percentile, whiskers minimum and maximum values. Virus is cleared significantly faster in secondary dengue compared to primary dengue (p = 0.002).
Fig 4. PK profile of castanospermine (semi-log…
Fig 4. PK profile of castanospermine (semi-log plot).
Solid brown line is the predicted concentration of castanospermine based on the mean PK parameters and the dosing regimen studied in the trial. Gray open circles are the observed peak and trough concentrations of castanospermine. Symbols and error bars are the mean and SEM, respectively. Black dotted line is the target trough concentration (400 ng/mL) predicted based on animal efficacy studies.
Fig 5. Dependence of pharmacokinetic parameters on…
Fig 5. Dependence of pharmacokinetic parameters on covariates.
Body Weight (A and B); Age (C and D); Creatinine Clearance (E); and Sex (F). Clearance or volume of distribution were not significantly affected by patients’ body weight, age or sex. Drug clearance was significantly correlated with creatinine clearance, indicating a significant role of the kidneys for elimination of celgosvir. Solid line-linear regression, dashed line- 95% CI. The slope of the linear regression line of creatinine clearance versus drug clearance was 0.86 (95% CI: 0.376, 1.351).
Fig 6. Scatterplots of drug exposure (Cmin,…
Fig 6. Scatterplots of drug exposure (Cmin, Cmax, AUC) and VLR2-4.
Blue open squares–individual patients who received placebo; red filled circles–individual patients who received celgosivir, separated into two quantiles of exposure. solid heavy line–median value, error bars– 25th to 75th percentile.
Fig 7. Predicted exposure for different dosing…
Fig 7. Predicted exposure for different dosing regimens.
The Box-25th to 75th percentile, whiskers-minimum and maximum values for the various dosing regimens is shown. (A) Cmin range for the various dosing regimens shows that 150 mg every 6 hr is predicted to yield a 4.5-fold increase in median Cmin used in CELADEN trial (B) Cmax, range do not vary significantly for the various dosing regimens and (C) AUC only shows a modest 1.33-fold increase over the dosing regimen used in the CELADEN trial.
Fig 8. Changes in platelets and hematocrit…
Fig 8. Changes in platelets and hematocrit for celgosivir (red filled circles) or placebo (blue open squares) treated patients.
Mean ± SEM changes in platelets count (A) and hematocrit (B) at different study days in all patients. (C) and (D) are Mean ± SEM changes in platelets count (C) and hematocrit (D) at different study days for secondary dengue patients only. Platelet nadir values (E) and difference between maximum and minimum hematocrit values (F) for secondary dengue patients treated with celgosivir or placebo, solid line–median, bars—interquartile range.
Fig 9. Celgosivir treatment changes the systemic…
Fig 9. Celgosivir treatment changes the systemic immune response during acute dengue infection.
A) Scatter plot of cytokine concentration of patient plasma samples drawn at specific time points post admission were analyzed for 41 cytokines and chemokines using the Human cytokine panel 1. The standard for each analyte between 4–10000 pg/ml is presented as yellow dashes, quality control for each analyte is in blue filled circles, a healthy volunteer (JEC) sample is shown in black filled circles the patient samples (400 samples/analyte; 8 per patient x 50 patients) are shown as green crosses. B) Heat map of log2 fold changes of averaged analyte concentrations at specific time points against the pre-treatment concentrations for selected analytes showing qualitative shifts in the analyte concentrations. Treatment and control groups fold changes are shown in separate rows for each analyte to aid comparison. Fold changes are colored from over expression in red to under expression in green. A scale showing the value of the maximum and minimum log2 fold change is shown.
Fig 10. Evolutionary relationship of the whole…
Fig 10. Evolutionary relationship of the whole genome of dengue samples isolated from celgosivir treatment and placebo in relation to representative samples collected from Asia.
Maximum likelihood trees were generated using the nucleotide alignment from start to stop codon of the coding region. Tip labels are coloured based on sample type, and genotypes are labelled adjacent to tip labels for DENV 2, and along branches for DENV 1 and 3. Scale bar represents nucleotide substitutions per site.

References

    1. Bhatt S, Gething PW, Brady OJ, Messina JP, Farlow AW, Moyes CL, et al. The global distribution and burden of dengue. Nature. 2013;496(7446):504–7. 10.1038/nature12060
    1. WHO. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. WHO Guidelines Approved by the Guidelines Review Committee. Geneva2009.
    1. Lee KS, Lo S, Tan SS, Chua R, Tan LK, Xu H, et al. Dengue virus surveillance in Singapore reveals high viral diversity through multiple introductions and in situ evolution. Infect Genet Evol. 2012;12(1):77–85. 10.1016/j.meegid.2011.10.012
    1. Communicable Diseases Surveillance in Singapore 2013. [accessed 01 Feb 2016].
    1. Wilder-Smith A, Ooi EE, Vasudevan SG, Gubler DJ. Update on dengue: epidemiology, virus evolution, antiviral drugs, and vaccine development. Curr Infect Dis Rep. 2010;12(3):157–64. 10.1007/s11908-010-0102-7
    1. Guzman MG, Alvarez M, Halstead SB. Secondary infection as a risk factor for dengue hemorrhagic fever/dengue shock syndrome: an historical perspective and role of antibody-dependent enhancement of infection. Arch Virol. 2013;158(7):1445–59. 10.1007/s00705-013-1645-3
    1. Halstead SB, O'Rourke EJ. Antibody-enhanced dengue virus infection in primate leukocytes. Nature. 1977;265(5596):739–41.
    1. Endy TP, Nisalak A, Chunsuttitwat S, Vaughn DW, Green S, Ennis FA, et al. Relationship of preexisting dengue virus (DV) neutralizing antibody levels to viremia and severity of disease in a prospective cohort study of DV infection in Thailand. J Infect Dis. 2004;189(6):990–1000.
    1. Vaughn DW, Green S, Kalayanarooj S, Innis BL, Nimmannitya S, Suntayakorn S, et al. Dengue viremia titer, antibody response pattern, and virus serotype correlate with disease severity. J Infect Dis. 2000;181(1):2–9.
    1. Capeding MR, Tran NH, Hadinegoro SR, Ismail HI, Chotpitayasunondh T, Chua MN, et al. Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. Lancet. 2014;384(9951):1358–65. 10.1016/S0140-6736(14)61060-6
    1. Villar L, Dayan GH, Arredondo-Garcia JL, Rivera DM, Cunha R, Deseda C, et al. Efficacy of a tetravalent dengue vaccine in children in Latin America. N Engl J Med. 2015;372(2):113–23. 10.1056/NEJMoa1411037
    1. Hadinegoro SR, Arredondo-Garcia JL, Capeding MR, Deseda C, Chotpitayasunondh T, Dietze R, et al. Efficacy and Long-Term Safety of a Dengue Vaccine in Regions of Endemic Disease. N Engl J Med. 2015;373(13):1195–206. 10.1056/NEJMoa1506223
    1. Halstead SB, Russell PK. Protective and immunological behavior of chimeric yellow fever dengue vaccine. Vaccine. 2016;34(14): 1643–47. 10.1016/j.vaccine.2016.02.004
    1. Tricou V, Minh NN, Van TP, Lee SJ, Farrar J, Wills B, et al. A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese adults. PLoS Negl Trop Dis. 2010;4(8):e785 10.1371/journal.pntd.0000785
    1. Nguyen NM, Tran CN, Phung LK, Duong KT, Huynh Hle A, Farrar J, et al. A randomized, double-blind placebo controlled trial of balapiravir, a polymerase inhibitor, in adult dengue patients. J Infect Dis. 2013;207(9):1442–50. 10.1093/infdis/jis470
    1. Tam DT, Ngoc TV, Tien NT, Kieu NT, Thuy TT, Thanh LT, et al. Effects of short-course oral corticosteroid therapy in early dengue infection in Vietnamese patients: a randomized, placebo-controlled trial. Clin Infect Dis. 2012;55(9):1216–24. 10.1093/cid/cis655
    1. Low JG, Sung C, Wijaya L, Wei Y, Rathore AP, Watanabe S, et al. Efficacy and safety of celgosivir in patients with dengue fever (CELADEN): a phase 1b, randomised, double-blind, placebo-controlled, proof-of-concept trial. Lancet Infect Dis. 2014;14(8):706–15. 10.1016/S1473-3099(14)70730-3
    1. Whitehorn J, Nguyen CV, Khanh LP, Kien DT, Quyen NT, Tran NT, et al. Lovastatin for the Treatment of Adult Patients With Dengue: A Randomized, Double-Blind, Placebo-Controlled Trial. Clin Infect Dis. 2016;62(4):468–76. 10.1093/cid/civ949
    1. Durantel D. Celgosivir, an alpha-glucosidase I inhibitor for the potential treatment of HCV infection. Curr Opin Investig Drugs. 2009;10(8):860–70.
    1. Sorbera LA, Castaner J, Garcia-Capdevilla L Celgosivir: α-Glucosidase inhibitor anti-hepatitis C virus drug. Drugs of the future 2005;30(6): 545–52.
    1. Rathore AP, Paradkar PN, Watanabe S, Tan KH, Sung C, Connolly JE, et al. Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-survival genes and protects against lethal challenge mouse model. Antiviral Res. 2011;92(3):453–60. 10.1016/j.antiviral.2011.10.002
    1. Watanabe S, Rathore AP, Sung C, Lu F, Khoo YM, Connolly J, et al. Dose- and schedule-dependent protective efficacy of celgosivir in a lethal mouse model for dengue virus infection informs dosing regimen for a proof of concept clinical trial. Antiviral Res. 2012;96(1):32–5. 10.1016/j.antiviral.2012.07.008
    1. Watanabe S, Chan KW, Dow G, Ooi EE, Low JG, Vasudevan SG. Optimizing celgosivir therapy in mouse models of dengue virus infection of serotypes 1 and 2: The search for a window for potential therapeutic efficacy. Antiviral Res. 2016;127:10–9. 10.1016/j.antiviral.2015.12.008
    1. Liu PS, Rhinehart BL, Daniel JK. Castanospermine Esters and Glycosides. US Patent 5,017,563. 1991.
    1. Kraus AA, Messer W, Haymore LB, de Silva AM. Comparison of plaque- and flow cytometry-based methods for measuring dengue virus neutralization. J Clin Microbiol. 2007;45(11):3777–80
    1. Chaterji S., Allen JC. Jr., Chow A, Leo YS, Ooi EE Evaluation of the NS1 rapid test and the WHO dengue classification schemes for use as bedside diagnosis of acute dengue fever in adults. Am J Trop Med Hyg, 2011. 84(2): p. 224–8. 10.4269/ajtmh.2011.10-0316
    1. Sessions OM, Wilm A, Kamaraj US, Choy MM, Chow A, Chong Y, et al. Analysis of Dengue Virus Genetic Diversity during Human and Mosquito Infection Reveals Genetic Constraints. PLoS Negl Trop Dis. 2015;9(9):e0004044 10.1371/journal.pntd.0004044
    1. Sim S, Aw PP, Wilm A, Teoh G, Hue KD, Nguyen NM, et al. Tracking Dengue Virus Intra-host Genetic Diversity during Human-to-Mosquito Transmission. PLoS Negl Trop Dis. 2015;9(9):e0004052 10.1371/journal.pntd.0004052
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60. 10.1093/bioinformatics/btp324
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303. 10.1101/gr.107524.110
    1. Wilm A, Aw PP, Bertrand D, Yeo GH, Ong SH, Wong CH, et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets, Nucleic Acids Res. 2012:40 (22): 11189–201. 10.1093/nar/gks918
    1. Thai KT, Henn MR, Zody MC, Tricou V, Nguyet NM, Charlebois P, et al. High-resolution analysis of intrahost genetic diversity in dengue virus serotype 1 infection identifies mixed infections. Journal of virology. 2012;86(2):835–43. 10.1128/JVI.05985-11
    1. Katoh K., Standley D.M., 2013. MAFFT Multiple Sequence Alignment Software Version 7: Improvements in Performance and Usability. Molecular biology and evolution 30, 772–780 10.1093/molbev/mst010
    1. Stamatakis A., 2014. RAxML Version 8: A tool for Phylogenetic Analysis and Post- Analysis of Large Phylogenies. Bioinformatics 30: 1312–1313. 10.1093/bioinformatics/btu033
    1. Beatty PR, Puerta-Guardo H, Killingbeck SS, Glasner DR, Hopkins K, Harris E. Dengue virus NS1 triggers endothelial permeability and vascular leak that is prevented by NS1 vaccination. Sci Transl Med. 2015;7(304):304ra141 10.1126/scitranslmed.aaa3787
    1. Modhiran N, Watterson D, Muller DA, Panetta AK, Sester DP, Liu L, et al. Dengue virus NS1 protein activates cells via Toll-like receptor 4 and disrupts endothelial cell monolayer integrity. Sci Transl Med. 2015;7(304):304ra142 10.1126/scitranslmed.aaa3863
    1. Kang W, Li Y, Zhuang Y, Zhao K, Huang D, Sun Y. Dynamic analysis of Th1/Th2 cytokine concentration during antiretroviral therapy of HIV-1/HCV co-infected patients. BMC Infect Dis. 2012,12, 102 10.1186/1471-2334-12-102
    1. Warfield KL, Plummer EM, Sayce AC, Alonzi DS, Tang W, Tyrrell BE, et al. Inhibition of endoplasmic reticulum glucosidases is required for in vitro and in vivo dengue antiviral activity by the iminosugar UV-4. Antiviral Res. 2016. March 2 10.1016/j.antiviral.2016.03.001
    1. Pharmacokinetics in Patients with Impaired Renal Function–Study Design, Data Analysis, and Impact on Dosing and Labeling, FDA Draft Guidance for Industry, March 2010.
    1. Tricou V, Minh NN, Farrar J, Tran HT, Simmons CP. Kinetics of viremia and NS1 antigenemia are shaped by immune status and virus serotype in adults with dengue. PLoS Negl Trop Dis. 2011;5(9):e1309 10.1371/journal.pntd.0001309
    1. Keller TH, Chen YL, Knox JE, et al. Finding new medicines for flaviviral targets. Novartis Foundation symposium 2006; 277:102–14; discussion 14–9, 251–3.
    1. World Health Organization. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control: New Edition. WHO Guidelines Approved by the Guidelines Review Committee. Geneva, 2009.
    1. Manokaran G, McPherson K, Simmons CP Stopping dengue: recent advances and new challenges. 2016. Microbiology Australia
    1. Sayce AC, Alonzi DS, Killingbeck SS, Tyrrell BE, Hill ML, et al. Iminosugras inhibit dengue virus production via inhibition of endoplasmic reticulum glucosidases–not glycolipid processing enzymes. 2016. PLoS Negl Trop Dis. 10.1371/journal.pntd.0004524
    1. Weaver SC, Costa F, Garcia-Blanco MA, Ko AI, Guilherme SR et al. Zika virus: History, Emergence, Biology and Prospects for Control. 2016 Antiviral Res. Antiviral Res. 2016. June;130:69–80.

Source: PubMed

3
Prenumerera