Safety, tolerability, and immune-biomarker profiling for year-long sargramostim treatment of Parkinson's disease

Katherine E Olson, Krista L Namminga, Yaman Lu, Aaron D Schwab, Mackenzie J Thurston, Mai M Abdelmoaty, Vikas Kumar, Melinda Wojtkiewicz, Helen Obaro, Pamela Santamaria, R Lee Mosley, Howard E Gendelman, Katherine E Olson, Krista L Namminga, Yaman Lu, Aaron D Schwab, Mackenzie J Thurston, Mai M Abdelmoaty, Vikas Kumar, Melinda Wojtkiewicz, Helen Obaro, Pamela Santamaria, R Lee Mosley, Howard E Gendelman

Abstract

Background: Neuroinflammation plays a pathogenic role in Parkinson's disease (PD). Immunotherapies that restore brain homeostasis can mitigate neurodegeneration by transforming T cell phenotypes. Sargramostim has gained considerable attention as an immune transformer through laboratory bench to bedside clinical studies. However, its therapeutic use has been offset by dose-dependent adverse events. Therefore, we performed a reduced drug dose regimen to evaluate safety and to uncover novel disease-linked biomarkers during 5 days/week sargramostim treatments for one year.

Methods: Five PD subjects were enrolled in a Phase 1b, unblinded, open-label study to assess safety and tolerability of 3 μg/kg/day sargramostim. Complete blood counts and chemistry profiles, physical examinations, adverse events (AEs), immune profiling, Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) scores, T cell phenotypes/function, DNA methylation, and gene and protein patterns were evaluated.

Findings: Sargramostim administered at 3 μg/kg/day significantly reduced numbers and severity of AEs/subject/month compared to 6 μg/kg/day treatment. While MDS-UPDRS Part III score reductions were recorded, peripheral blood immunoregulatory phenotypes and function were elevated. Hypomethylation of upstream FOXP3 DNA elements was also increased.

Interpretation: Long-term sargramostim treatment at 3 μg/kg/day is well-tolerated and effective in restoring immune homeostasis. There were decreased numbers and severity of AEs and restored peripheral immune function coordinate with increased numbers and function of Treg. MDS-UPDRS Part III scores did not worsen. Larger patient numbers need be evaluated to assess conclusive drug efficacy (ClinicalTrials.gov NCT03790670).

Funding: The research was supported by community funds to the University of Nebraska Foundation and federal research support from 5 R01NS034239-25.

Keywords: Granulocyte macrophage-colony stimulating factor; Neuroprotection; Parkinson's disease; Regulatory T cells; Sargramostim; Unified Parkinson's Disease Rating Scale.

Conflict of interest statement

Declaration of Competing Interest The authors declare no conflicts of interest.

Published by Elsevier B.V.

Figures

Fig. 1
Fig. 1
Adverse events (AE) comparing two clinical trials of sargramostim. PD subjects were administered sargramostim (Leukine®, human recombinant GM-CSF) in a previous Phase 1a (Ph 1a) (n = 10) and current Phase 1b (Ph 1b) clinical trial (n = 5). Subjects in the Ph 1a trial received 6 μg/kg of sargramostim every day for 2 months. In a proof-of-concept study to attenuate AE frequency and severity and extend administration, subjects in the Ph 1b trial received 3 μg/kg sargramostim on a 5 day on/2 day off regimen for 12 months. (a) Total number of adverse events (AEs) per subject recorded during the 2- and 12- month interventional period (Total) normalized on a monthly basis (Total/Mo). (b) AE severity scored on a scale of 1–3 severity as (1) mild, (2) moderate, or (3) severe. Mild events cause minimal discomfort or concern, may require minimal or no treatment, and do not interfere with daily activities. Moderate events were defined as causing discomfort, inconvenience, or concerns which were ameliorated with simple therapeutic measures. Severe adverse events were defined as causing discomfort or incapacitation that require prescription drug therapy or other treatments or interventions by medical personnel. Differences in means (± SD) for Total AE/Subject, Total AE/Subject/Mo, and Severity of AEs between Ph Ia vs Ph 1b trials were determined by Student's t-test and p values annotated above the pair-wise comparisons. (c) Graphical representation displaying reported AE based on AE severity (y axis), AE category (z axis), and day of treatment (x axis) for the Ph 1a trial. (d) Graphical representation displaying reported AE based on AE severity (y axis), AE category (z axis), and day of treatment (x axis) for the Ph 1b study. (c and d) AE categories are defined in Table 2.
Fig. 2
Fig. 2
MDS-Unified Parkinson's Disease Rating Scale (UPDRS) Part III motor assessment before and during sargramostim treatment. Prior to treatment, subjects (n = 5) underwent at least three separate baseline evaluations (at month -4, -3, -2, and/or -1 before initiation) and then began drug administration (3 ug/kg per day, 5 days on/2 days off). After treatment initiation, subjects were evaluated by the study neurologist once/month for 6 months and once/ every 2 months thereafter for 12 months. (a) Raw UPDRS Part III scores over time grouped for individual subjects (2001, 2003, 2004, 2005, 2006). (b) Total mean UPDRS Part III scores grouped by time of treatment. Blue dashed lines indicate mean baseline measurement. (c) Mean UPDRS Part III scores ± SD grouped by combined (All) and individual subjects. Specific p values are indicated above each subject. Baseline values are represented as blue circles and sargramostim treatment is represented as red squares. (d) Change from baseline in UPDRS Part III scores over time grouped for individual subjects. (e) Mean change from baseline ± SD in UPDRS Part III scores grouped by time of treatment. (f) Mean change from baseline in UPDRS Part III scores ± SD grouped by combined (All) and individual subjects. Specific p values are indicated above each subject. Baseline values are represented as blue circles and sargramostim treatment is represented as red squares. Significant differences (± SD) in baseline and treated means were determined by Student's t-test with p values denoted above comparisons. Differences in means ± SD over time were also determined by one-way ANOVA where p ≤ 0.05 compared to baseline (b).
Fig. 3
Fig. 3
Flow cytometric analysis of CD4+ peripheral blood populations over time. Quantification of frequencies for the following dependent variables: (a) CD4+ lymphocytes, (b) CD4+CD127lowCD25+ Tregs, (c) CD4+CD127highCD25+ Teffs, (d) CD4+FOXP3+, (e) CD4+FOXP3+HELIOS+, (f) CD4+CD31+, (g) CD4+CTLA+, (h) CD4+ItgB7+, (i) CD4+ItgA4B7+ over the course of treatment. Differences in means (± SD) for each dependent variable grouped by time on treatment were determined by one-way ANOVA and Dunnett's post hoc test where p ≤ 0.05 compared to baseline (*).
Fig. 4
Fig. 4
Flow cytometric analysis, immunosuppressive function, and FOXP3 Treg-Specific Demethylated Region (TSDR) methylation status of CD4+CD127lowCD25+ Treg subsets within CD4+ peripheral blood lymphocytes. Quantification for the dependent variables included (a) CD45RA-RO+ Treg, (b) CD31+ Treg, (c) CTLA+ Treg, (d) CD49+ Treg, and (e) ItgB7+ Treg subset frequencies within peripheral blood over time. Difference in means (± SD) for each dependent variable grouped by time of treatment were determined by one-way ANOVA and Dunnett's post hoc test where p ≤ 0.05 compared to baseline (*). (f) Percent demethylation (± SD) within the TSDR of the FOXP3 intron from isolated Tregs before and at 2 and 6 months after initiation of sargramostim treatment. Differences in means (± SD) were determined by one-way ANOVA where p ≤ 0.05 compared to baseline (*). (g) Quantification of Treg-mediated suppression (± SD) of Tresp (CD4+CD25-) proliferation at various Tresp:Treg ratios. Treg-mediated suppression is calculated as % Inhibition = 1 – (% Proliferating Tresp:Treg ÷ % Proliferating Stimulated Tresp Alone) and is reported as percent inhibition. Linear regression analysis indicates r2 ≥ 0.81, p < 0.0001 for all lines and significant elevation (p < 0.0001) from baseline (blue) compared to each month of treatment. (h) Correlation analysis for percent TSDR demethylation and corresponding Treg-mediated inhibition (Treg activity, AUC) at baseline (blue), 2-month (red), and 6-month (green) during sargramostim treatment, indicating a direct correlation of TSDR demethylation and Treg activity with r = 0.3212, p = 0.0004.
Fig. 5
Fig. 5
Elevated peripheral blood markers are associated with enhanced Treg function. Correlation analyses are depicted for the dependent variables including AUC for Treg activity and (a) %Integrin B7+CD4+ T cells, (b) %FOXP3+CD4+ T cells, (c) %FAS+CD4+ T cells, (d) %CD45RA-RO+CD4+ T cells, (e) %CD27+CCR7- Treg, (f) %Integrin B7+ Treg, (g) %Integrin A4B7+ Treg, and (h) %CD45RA-CD27+CCR7- Treg. For all correlation analyses, regression bands are indicated by dashed lines that encompass the 95% confidence intervals (red) and 95% prediction values (blue). Pearson r and p values are depicted on individual graphs. Data are displayed as scatter plots using the percentage of peripheral blood marker in either the total CD4 population or the Treg population against Treg activity, AUC. Correlations were determined using Pearson product–moment correlation coefficients, p values determined for correlation coefficients greater than 0.25, and the resulting 16 determinations adjusted for FDR. Best-fit lines were determined by linear regression.
Fig. 6
Fig. 6
Elevated peripheral blood markers and enhanced Treg function are associated with decreased UPDRS Part III scores. Correlation analyses for the dependent variables that included UPRS Part III scores and (a) %FAS+ CD4+ T cells, (b) %Integrin B7+ Tregs, (c) %CD45RA+CD27+CCR7- Tregs, (d) %CD27+CCR7- Tregs, and (e) Treg activity as determined by 50% Inhibitory Treg number. For all correlation analyses, regression bands are indicated by dashed lines that encompass the 95% confidence intervals (red) and 95% prediction values (blue). The Pearson r and p values are depicted on individual graphs. Data are displayed as scatter plots using the percentage of peripheral blood marker in total CD4 population, Treg population, or Treg function as determined about 50% inhibitory Treg number against UPDRS Part III scores. Correlations were determined using Pearson product-moment correlation coefficients, p values determined for correlation coefficients greater than 0.25, and the resulting 39 determinations adjusted for FDR. Best-fit lines were determined using linear regression.
Fig. 7
Fig. 7
Differential proteomic analysis of peripheral blood lymphocytes at 2 and 6 months after treatment. Volcano plots showing the fold change (treatment versus baseline) plotted against the p value highlighting significantly changed proteins (red – upregulation and green – downregulation; p ≤ 0.05 and an absolute fold change of 2). The vertical lines correspond to the absolute fold change of 2, and the horizontal line represents a p value of 0.05.

References

    1. Schwab A.D., Thurston M.J., Machhi J. Immunotherapy for Parkinson's disease. Neurobiol Dis. 2020;137
    1. Olanow C.W., Kieburtz K., Schapira A.H. Why have we failed to achieve neuroprotection in Parkinson's disease? Ann Neurol. 2008;64(Suppl 2):S101–S110.
    1. Guzman-Martinez L., Maccioni R.B., Andrade V., Navarrete L.P., Pastor M.G., Ramos-Escobar N. Neuroinflammation as a common feature of neurodegenerative disorders. Front Pharmacol. 2019;10:1008.
    1. Machhi J., Kevadiya B.D., Muhammad I.K. Harnessing regulatory T cell neuroprotective activities for treatment of neurodegenerative disorders. Mol Neurodegener. 2020;15(1):32.
    1. Kosloski L.M., Kosmacek E.A., Olson K.E., Mosley R.L., Gendelman H.E. GM-CSF induces neuroprotective and anti-inflammatory responses in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxicated mice. J Neuroimmunol. 2013;265(1-2):1–10.
    1. Olson K.E., Kosloski-Bilek L.M., Anderson K.M. Selective VIP receptor agonists facilitate immune transformation for dopaminergic neuroprotection in MPTP-intoxicated mice. J Neurosci. 2015;35(50):16463–16478.
    1. Reynolds A.D., Banerjee R., Liu J., Gendelman H.E., Mosley R.L. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson's disease. J Leukoc Biol. 2007;82(5):1083–1094.
    1. Saunders J.A., Estes K.A., Kosloski L.M. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. J Neuroimmune Pharmacol. 2012;7(4):927–938.
    1. Chao Y., Wong S.C., Tan E.K. Evidence of inflammatory system involvement in Parkinson's disease. Biomed Res Int. 2014;2014
    1. Lindestam Arlehamn C.S., Dhanwani R., Pham J. Alpha-synuclein-specific T cell reactivity is associated with preclinical and early Parkinson's disease. Nat Commun. 2020;11(1):1875.
    1. Kustrimovic N., Comi C., Magistrelli L. Parkinson's disease patients have a complex phenotypic and functional Th1 bias: cross-sectional studies of CD4+ Th1/Th2/T17 and Treg in drug-naive and drug-treated patients. J Neuroinflammation. 2018;15(1):205.
    1. Olson K.E., Namminga K.L., Schwab A.D. Neuroprotective activities of long-acting granulocyte-macrophage colony-stimulating factor (mPDM608) in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-intoxicated mice. Neurotherapeutics. 2020;17(4):1861–1877.
    1. Gendelman H.E., Zhang Y., Santamaria P. Evaluation of the safety and immunomodulatory effects of sargramostim in a randomized, double-blind phase 1 clinical Parkinson's disease trial. NPJ Parkinsons Dis. 2017;3:10.
    1. Benjamini Y., Krieger A.M., Yekutieli D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika. 2006;93:491–507.
    1. Sulzer D., Alcalay R.N., Garretti F. T cells from patients with Parkinson's disease recognize alpha-synuclein peptides. Nature. 2017;546(7660):656–661.
    1. Baldo B.A. Side effects of cytokines approved for therapy. Drug Saf. 2014;37(11):921–943.
    1. Holden S.K., Finseth T., Sillau S.H., Berman B.D. Progression of MDS-UPDRS scores over five years in de novo Parkinson disease from the Parkinson's progression markers initiative cohort. Mov Disord Clin Pract. 2018;5(1):47–53.
    1. Evers L.J.W., Krijthe J.H., Meinders M.J., Bloem B.R., Heskes T.M. Measuring Parkinson's disease over time: The real-world within-subject reliability of the MDS-UPDRS. Mov Disord. 2019;34(10):1480–1487.
    1. Quattrone A., Barbagallo G., Cerasa A., Stoessl A.J. Neurobiology of placebo effect in Parkinson's disease: what we have learned and where we are going. Mov Disord. 2018;33(8):1213–1227.
    1. DeNucci C.C., Pagan A.J., Mitchell J.S., Shimizu Y. Control of alpha4beta7 integrin expression and CD4 T cell homing by the beta1 integrin subunit. J Immunol. 2010;184(5):2458–2467.
    1. Stassen M., Fondel S., Bopp T. Human CD25+ regulatory T cells: two subsets defined by the integrins alpha 4 beta 7 or alpha 4 beta 1 confer distinct suppressive properties upon CD4+ T helper cells. Eur J Immunol. 2004;34(5):1303–1311.
    1. Swerlick R.A., Lee K.H., Li L.J., Sepp N.T., Caughman S.W., Lawley T.J. Regulation of vascular cell adhesion molecule 1 on human dermal microvascular endothelial cells. J Immunol. 1992;149(2):698–705.
    1. Kanwar J.R., Harrison J.E., Wang D. Beta7 integrins contribute to demyelinating disease of the central nervous system. J Neuroimmunol. 2000;103(2):146–152.
    1. Sun H., Kuk W., Rivera-Nieves J., Lopez-Ramirez M.A., Eckmann L., Ginsberg M.H. Beta7 integrin inhibition can increase intestinal inflammation by impairing homing of CD25(hi)FoxP3(+) regulatory T cells. Cell Mol Gastroenterol Hepatol. 2020;9(3):369–385.
    1. Klann J.E., Kim S.H., Remedios K.A. Integrin activation controls regulatory T cell-mediated peripheral tolerance. J Immunol. 2018;200(12):4012–4023.
    1. Lehmann J., Huehn J., de la Rosa M. Expression of the integrin alpha Ebeta 7 identifies unique subsets of CD25+ as well as CD25- regulatory T cells. Proc Natl Acad Sci U S A. 2002;99(20):13031–13036.
    1. Fan X., Moltedo B., Mendoza A. CD49b defines functionally mature Treg cells that survey skin and vascular tissues. J Exp Med. 2018;215(11):2796–2814.
    1. Huang L., Zheng Y., Yuan X. Decreased frequencies and impaired functions of the CD31(+) subpopulation in Treg cells associated with decreased FoxP3 expression and enhanced Treg cell defects in patients with coronary heart disease. Clin Exp Immunol. 2017;187(3):441–454.
    1. Haas J., Korporal M., Balint B., Fritzsching B., Schwarz A., Wildemann B. Glatiramer acetate improves regulatory T-cell function by expansion of naive CD4(+)CD25(+)FOXP3(+)CD31(+) T-cells in patients with multiple sclerosis. J Neuroimmunol. 2009;216(1-2):113–117.
    1. Lu L., Barbi J., Pan F. The regulation of immune tolerance by FOXP3. Nat Rev Immunol. 2017;17(11):703–717.
    1. Shevyrev D., Tereshchenko V. Treg heterogeneity, function, and homeostasis. Front Immunol. 2019;10:3100.
    1. Walker L.S. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun. 2013;45:49–57.
    1. Wing J.B., Ise W., Kurosaki T., Sakaguchi S. Regulatory T cells control antigen-specific expansion of Tfh cell number and humoral immune responses via the coreceptor CTLA-4. Immunity. 2014;41(6):1013–1025.
    1. Borsellino G., Kleinewietfeld M., Di Mitri D. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood. 2007;110(4):1225–1232.
    1. Deaglio S., Dwyer K.M., Gao W. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204(6):1257–1265.
    1. Gu J., Ni X., Pan X. Human CD39(hi) regulatory T cells present stronger stability and function under inflammatory conditions. Cell Mol Immunol. 2017;14(6):521–528.
    1. Stojakovic A., Paz-Filho G., Arcos-Burgos M., Licinio J., Wong M.L., Mastronardi C.A. Role of the IL-1 pathway in dopaminergic neurodegeneration and decreased voluntary movement. Mol Neurobiol. 2017;54(6):4486–4495.
    1. Borst J., Hendriks J., Xiao Y. CD27 and CD70 in T cell and B cell activation. Curr Opin Immunol. 2005;17(3):275–281.
    1. Menning A., Hopken U.E., Siegmund K., Lipp M., Hamann A., Huehn J. Distinctive role of CCR7 in migration and functional activity of naive- and effector/memory-like Treg subsets. Eur J Immunol. 2007;37(6):1575–1583.
    1. Cowan J.E., McCarthy N.I., Anderson G. CCR7 controls thymus recirculation, but not production and emigration, of Foxp3(+) T cells. Cell Rep. 2016;14(5):1041–1048.
    1. Booth N.J., McQuaid A.J., Sobande T. Different proliferative potential and migratory characteristics of human CD4+ regulatory T cells that express either CD45RA or CD45RO. J Immunol. 2010;184(8):4317–4326.
    1. Park M.Y., Lim B.G., Kim S.Y., Sohn H.J., Kim S., Kim T.G. GM-CSF promotes the expansion and differentiation of cord blood myeloid-derived suppressor cells, which attenuate xenogeneic graft-vs.-host disease. Front Immunol. 2019;10:183.
    1. Hamilton J.A., Achuthan A. Colony stimulating factors and myeloid cell biology in health and disease. Trends Immunol. 2013;34(2):81–89.
    1. Pulendran B., Banchereau J., Burkeholder S. Flt3-ligand and granulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo. J Immunol. 2000;165(1):566–572.
    1. Bhattacharya P., Thiruppathi M., Elshabrawy H.A., Alharshawi K., Kumar P., Prabhakar B.S. GM-CSF: an immune modulatory cytokine that can suppress autoimmunity. Cytokine. 2015;75(2):261–271.
    1. Sheng J.R., Quan S., Soliven B. CD1d(hi)CD5+ B cells expanded by GM-CSF in vivo suppress experimental autoimmune myasthenia gravis. J Immunol. 2014;193(6):2669–2677.
    1. Schutt C.R., Gendelman H.E., Mosley R.L. Tolerogenic bone marrow-derived dendritic cells induce neuroprotective regulatory T cells in a model of Parkinson's disease. Mol Neurodegener. 2018;13(1):26.
    1. Alvarez-Luquin D.D., Arce-Sillas A., Leyva-Hernandez J. Regulatory impairment in untreated Parkinson's disease is not restricted to Tregs: other regulatory populations are also involved. J Neuroinflammation. 2019;16(1):212.
    1. Beers D.R., Zhao W., Wang J. ALS patients' regulatory T lymphocytes are dysfunctional, and correlate with disease progression rate and severity. JCI Insight. 2017;2(5):e89530.
    1. Faridar A., Thome A.D., Zhao W. Restoring regulatory T-cell dysfunction in Alzheimer's disease through ex vivo expansion. Brain Commun. 2020;2(2):fcaa112.
    1. Schreiber L., Pietzsch B., Floess S. The Treg-specific demethylated region stabilizes Foxp3 expression independently of NF-kappaB signaling. PLoS One. 2014;9(2):e88318.
    1. Anderson M.R., Enose-Akahata Y., Massoud R. Epigenetic modification of the FoxP3 TSDR in HAM/TSP decreases the functional suppression of Tregs. J Neuroimmune Pharmacol. 2014;9(4):522–532.
    1. Shimazu Y., Shimazu Y., Hishizawa M. Hypomethylation of the Treg-specific demethylated region in FOXP3 Is a hallmark of the regulatory T-cell subtype in adult T-cell Leukemia. Cancer Immunol Res. 2016;4(2):136–145.
    1. Reynolds A.D., Stone D.K., Hutter J.A., Benner E.J., Mosley R.L., Gendelman H.E. Regulatory T cells attenuate Th17 cell-mediated nigrostriatal dopaminergic neurodegeneration in a model of Parkinson's disease. J Immunol. 2010;184(5):2261–2271.
    1. Caraveo G., Auluck P.K., Whitesell L. Calcineurin determines toxic versus beneficial responses to alpha-synuclein. Proc Natl Acad Sci U S A. 2014;111(34):E3544–E3552.
    1. Hermann-Kleiter N., Baier G. NFAT pulls the strings during CD4+ T helper cell effector functions. Blood. 2010;115(15):2989–2997.
    1. Hogan P.G., Chen L., Nardone J., Rao A. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev. 2003;17(18):2205–2232.
    1. Kipanyula M.J., Kimaro W.H., Seke Etet P.F. The emerging roles of the calcineurin-nuclear factor of activated T-lymphocytes pathway in nervous system functions and diseases. J Aging Res. 2016;2016
    1. Liu X., Jana M., Dasgupta S. Human immunodeficiency virus type 1 (HIV-1) tat induces nitric-oxide synthase in human astroglia. J Biol Chem. 2002;277(42):39312–39319.
    1. Dasgupta S., Jana M., Liu X., Pahan K. Role of very-late antigen-4 (VLA-4) in myelin basic protein-primed T cell contact-induced expression of proinflammatory cytokines in microglial cells. J Biol Chem. 2003;278(25):22424–22431.
    1. Nagatsu T., Mogi M., Ichinose H., Togari A. Changes in cytokines and neurotrophins in Parkinson's disease. J Neural Transm Suppl. 2000;(60):277–290.
    1. Mogi M., Harada M., Kondo T. Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming growth factor-alpha are elevated in the brain from parkinsonian patients. Neurosci Lett. 1994;180(2):147–150.
    1. Ghosh A., Roy A., Liu X. Selective inhibition of NF-kappaB activation prevents dopaminergic neuronal loss in a mouse model of Parkinson's disease. Proc Natl Acad Sci U S A. 2007;104(47):18754–18759.

Source: PubMed

3
Abonner