PF-04447943, a Phosphodiesterase 9A Inhibitor, in Stable Sickle Cell Disease Patients: A Phase Ib Randomized, Placebo-Controlled Study

Robert J Charnigo, David Beidler, Denis Rybin, Debra D Pittman, Beesan Tan, Jo Howard, Alan D Michelson, Andrew L Frelinger , III, Nicholas Clarke, Robert J Charnigo, David Beidler, Denis Rybin, Debra D Pittman, Beesan Tan, Jo Howard, Alan D Michelson, Andrew L Frelinger , III, Nicholas Clarke

Abstract

This phase Ib study randomized patients with stable sickle cell disease (SCD) aged 18-65 years to twice-daily PF-04447943 (a phosphodiesterase 9A inhibitor; 5 or 25 mg) or placebo, with/without hydroxyurea coadministration, for up to 29 days. Blood samples were collected at baseline and various posttreatment time points for assessments of PF-04447943 pharmacokinetics (PKs)/pharmacodynamics (PDs). Change from baseline in potential SCD-related biomarkers was evaluated. Of 30 patients, 15 received hydroxyurea and 28 completed the study. PF-04447943, with/without hydroxyurea, was generally well tolerated, with no treatment-related serious adverse events. Plasma PF-04447943 exposure was dose proportional. Twice-daily PF-04447943 25 mg significantly reduced the number and size of circulating monocyte-platelet and neutrophil-platelet aggregates and levels of circulating soluble E-selectin at day 29 vs. baseline (adjusted P < 0.15). PF-04447943 demonstrated PK/PD effects suggestive of inhibiting pathways that may contribute to vaso-occlusion. This study also provides guidance regarding biomarkers for future SCD studies.

Trial registration: ClinicalTrials.gov NCT02114203.

Conflict of interest statement

R.J.C., D.B., D.R., D.D.P., and N.C. are employees of Pfizer Inc and may own stock/options in the company. B.T. was an employee of Pfizer, Inc at the time of this study. J.H. is a consultant for Bluebird Bio, GBT, and Pfizer, and a member of the Speakers’ Bureau for Addmedica, Novartis, and Terumo BCT. A.D.M. has served as a consultant for Instrumentation Laboratory; receives research funding from Baxalta, Eisai, GLSynthesis, Ionis, Ironwood, Pfizer, and Sysmex; and is a member of advisory committees for AstraZeneca and Janssen. A.L.F. has received research funding from Baxalta, Eisai, GLSynthesis, Ionis, Ironwood, Pfizer, and Sysmex.

© 2018 Pfizer Inc. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Schematic of the study design. Cohort 1 received 25 mg PF‐04447943 or placebo twice a day for 29 days (n = 15,a 3:1 active‐placebo ratio). Cohort 2 received 5 mg PF‐04447943 or placebo twice a day for 29 days (n = 7, 3:1 active‐placebo ratio). CI, confidence interval; HU, hydroxyurea; ICAM, intercellular adhesion molecule; RBC, red blood cell; VCAM, vascular adhesion molecule. aOne patient discontinued after randomization (n=16) but before receiving study drug.
Figure 2
Figure 2
Percent change from baseline at day 29 in biomarkers with PF‐04447943 25 mg twice daily. The percent changes from baseline to day 29 in adhesion, aggregates, microparticle, coagulation, and hematology biomarkers are depicted in the forest plot. Estimates and 95% confidence intervals are based on repeated measures model. Statistically significant (< 0.15 after multiplicity correction using Holm‐Bonferroni stepdown procedure) changes from baseline are indicated by an asterisk. CI, confidence interval; ICAM, intercellular adhesion molecule; MPA, monocyte‐platelet aggregates; NPA, neutrophil‐platelet aggregate; RBC, red blood cell; TAT, thrombin‐antithrombin; VCAM, vascular adhesion molecule.
Figure 3
Figure 3
Percent change from baseline at day 29 in cellular aggregates and soluble P‐selectin and E‐selectin levels. (a) Change in monocyte‐platelet aggregates, number, and size. (b) Change in neutrophil‐platelet aggregates, number, and size. (c) Change in plasma‐soluble E‐selectin and P‐selectin. Bars and whiskers represent estimates and 95% confidence intervals (CIs), based on repeated measures model. The P values of statistically significant (< 0.15 after multiplicity correction using Holm‐Bonferroni stepdown procedure) changes from baseline are reported above corresponding estimates. MPA, monocyte‐platelet aggregates; NPA, neutrophil‐platelet aggregate.
Figure 4
Figure 4
Analysis of biomarkers by hydroxyurea (HU) use and fetal hemoglobin (HbF) levels. (a) Box and whiskers plot of HbF baseline levels (%) in patients (n = 27) with and without HU treatment. Boxes present area between 25th and 75th percentile values. Whiskers extend 1.5 of interquartile range from the box. Line within each box is the median value. The circles represent individual observations that could be considered outliers. (b) The percent changes from baseline at day 29 for biomarkers in subgroups of patients (PF‐04447943 25 mg twice daily), with vs. without HU cotreatment, and low vs. high baseline HbF levels. Low HbF levels were defined as < 10%, and high baseline HbF levels were 10% or higher, based on median of the sample distribution. Forest plot presents estimates and 95% confidence intervals, based on repeated measures model.

References

    1. Piel, F.B. , Steinberg, M.H. & Rees, D.C. Sickle cell disease. N. Engl. J. Med. 376, 1561–1573 (2017).
    1. Almeida, C.B. et al Hydroxyurea and a cGMP‐amplifying agent have immediate benefits on acute vaso‐occlusive events in sickle cell disease mice. Blood 120, 2879–2888 (2012).
    1. Hassell, K.L. Population estimates of sickle cell disease in the U.S. Am. J. Prev. Med. 38, S512–S521 (2010).
    1. Piel, F.B. , Hay, S.I. , Gupta, S. , Weatherall, D.J. & Williams, T.N. Global burden of sickle cell anaemia in children under five, 2010‐2050: modelling based on demographics, excess mortality, and interventions. PLoS Med. 10, e1001484 (2013).
    1. Hoover, R. , Rubin, R. , Wise, G. & Warren, R. Adhesion of normal and sickle erythrocytes to endothelial monolayer cultures. Blood 54, 872–876 (1979).
    1. Turhan, A. , Weiss, L.A. , Mohandas, N. , Coller, B.S. & Frenette, P.S. Primary role for adherent leukocytes in sickle cell vascular occlusion: a new paradigm. Proc. Natl. Acad. Sci. USA 99, 3047–3051 (2002).
    1. Belcher, J.D. et al Critical role of endothelial cell activation in hypoxia‐induced vasoocclusion in transgenic sickle mice. Am. J. Physiol. Heart Circ. Physiol. 288, H2715–H2725 (2005).
    1. Zennadi, R. , Chien, A. , Xu, K. , Batchvarova, M. & Telen, M.J. Sickle red cells induce adhesion of lymphocytes and monocytes to endothelium. Blood 112, 3474–3483 (2008).
    1. Frenette, P.S. & Atweh, G.F. Sickle cell disease: old discoveries, new concepts, and future promise. J. Clin. Invest. 117, 850–858 (2007).
    1. Zhang, D. , Xu, C. , Manwani, D. & Frenette, P.S. Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 127, 801–809 (2016).
    1. Powars, D.R. , Chan, L.S. , Hiti, A. , Ramicone, E. & Johnson, C. Outcome of sickle cell anemia: a 4‐decade observational study of 1056 patients. Medicine 84, 363–376 (2005).
    1. Platt, O.S. et al Pain in sickle cell disease. Rates and risk factors. N. Engl. J. Med. 325, 11–16 (1991).
    1. Darbari, D.S. et al Severe painful vaso‐occlusive crises and mortality in a contemporary adult sickle cell anemia cohort study. PLoS One 8, e79923 (2013).
    1. Canalli, A.A. , Franco‐Penteado, C.F. , Saad, S.T. , Conran, N. & Costa, F.F. Increased adhesive properties of neutrophils in sickle cell disease may be reversed by pharmacological nitric oxide donation. Haematologica 93, 605–609 (2008).
    1. Cokic, V.P. et al Hydroxyurea induces fetal hemoglobin by the nitric oxide‐dependent activation of soluble guanylyl cyclase. J. Clin. Invest. 111, 231–239 (2003).
    1. Kleiman, R.J. et al Phosphodiesterase 9A regulates central cGMP and modulates responses to cholinergic and monoaminergic perturbation in vivo. J. Pharmacol. Exp. Ther. 341, 396–409 (2012).
    1. Hutson, P.H. et al The selective phosphodiesterase 9 (PDE9) inhibitor PF‐04447943 (6‐[(3S,4S)‐4‐methyl‐1‐(pyrimidin‐2‐ylmethyl)pyrrolidin‐3‐yl]‐1‐(tetrahydro‐2H‐py ran‐4‐yl)‐1,5‐dihydro‐4H‐pyrazolo[3,4‐d]pyrimidin‐4‐one) enhances synaptic plasticity and cognitive function in rodents. Neuropharmacology 61, 665–676 (2011).
    1. Verhoest, P.R. et al Design and discovery of 6‐[(3S,4S)‐4‐methyl‐1‐(pyrimidin‐2‐ylmethyl)pyrrolidin‐3‐yl]‐1‐(tetrahydro‐2H‐pyr an‐4‐yl)‐1,5‐dihydro‐4H‐pyrazolo[3,4‐d]pyrimidin‐4‐one (PF‐04447943), a selective brain penetrant PDE9A inhibitor for the treatment of cognitive disorders. J. Med. Chem. 55, 9045–9054 (2012).
    1. Vardigan, J.D. , Converso, A. , Hutson, P.H. & Uslaner, J.M. The selective phosphodiesterase 9 (PDE9) inhibitor PF‐04447943 attenuates a scopolamine‐induced deficit in a novel rodent attention task. J. Neurogenet. 25, 120–126 (2011).
    1. Wu, L.C. , Sun, C.W. , Ryan, T.M. , Pawlik, K.M. , Ren, J. & Townes, T.M. Correction of sickle cell disease by homologous recombination in embryonic stem cells. Blood 108, 1183–1188 (2006).
    1. Jasuja, R. , Patel Hett, S. , Fruebis, J. & Pittman, D. PDE‐9 inhibition combined with hydroxyurea is beneficial in vaso‐occlusive crisis in mouse model of sickle cell disease [abstract]. Blood 124, 2694 (2014).
    1. Jasuja, R. , Parks, E. , Murphy, J.E. & Pittman, D.D. Chronic administration of the PDE9 inhibitor PF‐04447943 reduces leukocyte‐platelet aggregates and markers of endothelial activation in a mouse model of sickle cell disease [abstract]. Blood 128, 1293 (2016).
    1. Evans, R.M. et al Safety and pharmacokinetics of PF‐04447943, a PDEA inhibitor, in single and multiple dose phase 1 studies in healthy volunteers [abstract O3‐05‐04]. Alzheimers Dement. 6, S135 (2010).
    1. Nicholas, T. et al PF‐04447943, a novel PDE9A inhibitor, increases CGMP levels in cerebrospinal fluid: translation from non‐clinical species to healthy human volunteers [abstract P2‐240]. Alzheimers Dement. 5, P330–P331 (2009).
    1. Schwam, E.M. et al A multicenter, double‐blind, placebo‐controlled trial of the PDE9A inhibitor, PF‐04447943 Alzheimer's disease. Curr. Alzheimer Res. 11, 413–421 (2014).
    1. Michelson, A.D. , Barnard, M.R. , Krueger, L.A. , Frelinger, A.L. III & Furman, M.I. Evaluation of platelet function by flow cytometry. Methods 21, 259–270 (2000).
    1. Gerrits, A.J. , Frelinger, A.L. III & Michelson, A.D. Whole blood analysis of leukocyte‐platelet aggregates. Curr. Protoc. Cytom. 78, 6.15.1–6.15.10 (2016).
    1. R Core Team . R: a language and environment for statistical computing. R Foundation for Statistical Computing <>. (2016) Accessed: October 9, 2018.
    1. Xie, R. , Tan, B. & Harnisch, L.O . Population pharmacokinetics of PF‐04447943 in health volunteers and adult patients with Alzheimer's disease or sickle cell disease [poster]. Presented at: Annual Meeting of the Population Approach Group in Europe; June 6–9, 2017; Budapest, Hungary.
    1. Chang, J. , Patton, J.T. , Sarkar, A. , Ernst, B. , Magnani, J.L. & Frenette, P.S. GMI‐1070, a novel pan‐selectin antagonist, reverses acute vascular occlusions in sickle cell mice. Blood 116, 1779–1786 (2010).
    1. Wun, T. et al Phase 1 study of the E‐selectin inhibitor GMI 1070 in patients with sickle cell anemia. PLoS One 9, e101301 (2014).
    1. Telen, M.J. et al Randomized phase 2 study of GMI‐1070 in SCD: reduction in time to resolution of vaso‐occlusive events and decreased opioid use. Blood 125, 2656–2664 (2015).

Source: PubMed

3
Tilaa