Expression of γ-globin genes in β-thalassemia patients treated with sirolimus: results from a pilot clinical trial (Sirthalaclin)

Cristina Zuccato, Lucia Carmela Cosenza, Matteo Zurlo, Jessica Gasparello, Chiara Papi, Elisabetta D'Aversa, Giulia Breveglieri, Ilaria Lampronti, Alessia Finotti, Monica Borgatti, Chiara Scapoli, Alice Stievano, Monica Fortini, Eric Ramazzotti, Nicola Marchetti, Marco Prosdocimi, Maria Rita Gamberini, Roberto Gambari, Cristina Zuccato, Lucia Carmela Cosenza, Matteo Zurlo, Jessica Gasparello, Chiara Papi, Elisabetta D'Aversa, Giulia Breveglieri, Ilaria Lampronti, Alessia Finotti, Monica Borgatti, Chiara Scapoli, Alice Stievano, Monica Fortini, Eric Ramazzotti, Nicola Marchetti, Marco Prosdocimi, Maria Rita Gamberini, Roberto Gambari

Abstract

Introduction: β-thalassemia is caused by autosomal mutations in the β-globin gene, which induce the absence or low-level synthesis of β-globin in erythroid cells. It is widely accepted that a high production of fetal hemoglobin (HbF) is beneficial for patients with β-thalassemia. Sirolimus, also known as rapamycin, is a lipophilic macrolide isolated from a strain of Streptomyces hygroscopicus that serves as a strong HbF inducer in vitro and in vivo. In this study, we report biochemical, molecular, and clinical results of a sirolimus-based NCT03877809 clinical trial (a personalized medicine approach for β-thalassemia transfusion-dependent patients: testing sirolimus in a first pilot clinical trial, Sirthalaclin).

Methods: Accumulation of γ-globin mRNA was analyzed using reverse-transcription quantitative polymerase chain reaction (PCR), while the hemoglobin pattern was analyzed using high-performance liquid chromatography (HPLC). The immunophenotype was analyzed using a fluorescence-activated cell sorter (FACS), with antibodies against CD3, CD4, CD8, CD14, CD19, CD25 (for analysis of peripheral blood mononuclear cells), or CD71 and CD235a (for analysis of in vitro cultured erythroid precursors).

Results: The results were obtained in eight patients with the β+/β+ and β+/β0 genotypes, who were treated with a starting dosage of 1 mg/day sirolimus for 24-48 weeks. The first finding of this study was that the expression of γ-globin mRNA increased in the blood and erythroid precursor cells isolated from β-thalassemia patients treated with low-dose sirolimus. This trial also led to the important finding that sirolimus influences erythropoiesis and reduces biochemical markers associated with ineffective erythropoiesis (excess free α-globin chains, bilirubin, soluble transferrin receptor, and ferritin). A decrease in the transfusion demand index was observed in most (7/8) of the patients. The drug was well tolerated, with minor effects on the immunophenotype, and an only side effect of frequently occurring stomatitis.

Conclusion: The data obtained indicate that low doses of sirolimus modify hematopoiesis and induce increased expression of γ-globin genes in a subset of patients with β-thalassemia. Further clinical trials are warranted, possibly including testing of the drug in patients with less severe forms of the disease and exploring combination therapies.

Keywords: fetal hemoglobin; ineffective erythropoiesis; sirolimus; transfusion requirement; β-thalassemia; γ-globin mRNA.

Conflict of interest statement

Conflict of interest statement: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: M.P. is the administrator of ‘Rare Partners srl Impresa Sociale’ to whom the rights for the patent WO 2004/004697 on the use of sirolimus in β-thalassemia have been assigned.

© The Author(s), 2022.

Figures

Figure 1.
Figure 1.
Flowchart summarizing the NCT03877809 Sirthalaclin clinical trial and the key analysis presented in this study. Five patients concluded the trial at V8 (180 days), as indicated. HbF and excess of free α-globin chains were analyzed using HPLC. Content of γ-globin mRNA was analyzed using RT-qPCR. The scheme of the trial has been described elsewhere.
Figure 2.
Figure 2.
Representative HPLC patterns obtained after exposure of ErPCs isolated from patients (a) n.9, (b) n.14, (c) n.15, and (d) n.17 to sirolimus. The arrows indicate the positions of the HbA, HbA2, and HbF0 peaks.
Figure 3.
Figure 3.
Blood levels of sirolimus (ng/mL). For calculating the average values, the samples with sirolimus blood concentrations p-value for the comparison of V8 data to V3 data, samples from patient n.24 were not considered.
Figure 4.
Figure 4.
Increase in the content of γ-globin mRNA in the blood of patients treated with sirolimus (a–c). Representative data obtained from the blood samples of patient n.11 using (a) RPL13A, (b) GAPDH, and (c) β-actin control sequences, as indicated. (d) Increase in the content of γ-globin mRNA in the blood. The data represent fold values with respect to V2 of patient n.11. (e) Average increase values of γ-globin mRNA expressed as fold content with respect to V2 of patient n.11. Raw data are presented in Supplementary Table S5. N = 8 (V2–V8) and 3 (V11).
Figure 5.
Figure 5.
Increase in the content of γ-globin mRNA in the ErPCs isolated from patients treated with sirolimus. (a) Increase in the content of γ-globin mRNA in the ErPCs. The data represent fold values with respect to V2 of patient n.24. (b) Average increase in the content of γ-globin mRNA. The data represent fold values with respect to V2 of patient n.24. Raw data are presented in Supplementary Table S6. N = 8 (V2–V8) and 3 (V11).
Figure 6.
Figure 6.
Increase in percentage of HbF in EPO-induced ErPCs isolated at V6. (a) Representative HPLC analysis of EPO-treated ErPCs from patients n.11 and n.18 at V2 and V6 Sirthalaclin visits. The arrows indicate positions of the HbA, HbA2, and HbF peaks. (b) Changes in the percentage of HbF at V6 (black histograms) with respect to V2 (white histograms). The cumulative data from all the analyzed patients are also shown.
Figure 7.
Figure 7.
(a) Changes in free α-globin chains, (b) bilirubin, (c) sTFR, and (d) ferritin levels, following treatment with sirolimus. Only six patients were analyzed for changes of free α-globin chains, since two patients exhibited lack of detectable α-globin peak on HPLC analysis. Raw data related to panels B–D are presented in Supplementary Tables S7S9. N = 8 (V2–V8) and 3 (V11).
Figure 8.
Figure 8.
Changes in the transfusion demand (TD) when data at V2 and V8 are considered (p = 0.006). N = 8.
Figure 9.
Figure 9.
Immunophenotype of PBMCs from sirolimus-treated β-thalassemia patients: FACS analysis. A representative example of the gating strategy employed is given in Supplementary Figure S5A–C. The summary of the results obtained is shown focusing on the following markers associated with the various lymphocyte subpopulations: (a) CD14, (b) CD19, (c) CD3, (d) CD4, (e) CD8, and (f) CD25. With this panel of antibodies, we were able to subdivide the PBMCs population into monocytes (CD14+), B cells (CD14−/CD3−/CD19+), T cells (CD14−/CD3+), CD4+ T cells (CD14−/CD3+/CD4+), CD8+ T cells (CD14−/CD3+/CD8+), and CD8+ activated T cells (CD14−/CD3+/CD8+/CD25+). Average changes of the immunophenotype pattern at V2, V6, V8, and V11 are reported. N = 8 (V2–V8) and 3 (V11).

References

    1. Weatherall DJ. Phenotype-genotype relationships in monogenic disease: lessons from the thalassaemias. Nat Rev Genet 2001; 2: 245–255.
    1. Origa R. β-thalassemia. Genet Med 2017; 19: 609–619.
    1. Fucharoen S, Weatherall DJ. Progress toward the control and management of the thalassemias. Hematol Oncol Clin North Am 2016; 30: 359–371.
    1. Modell B, Darlison M, Birgens H, et al.. Epidemiology of haemoglobin disorders in Europe: an overview. Scand J Clin Lab Invest 2007; 67: 39–69.
    1. Galanello R, Origa R. B-thalassemia. Orphanet J Rare Dis 2010; 5: 11.
    1. Colah R, Gorakshakar A, Nadkarni A. Global burden, distribution and prevention of β-thalassemias and hemoglobin E disorders. Expert Rev Hematol 2010; 3: 103–117.
    1. Modell B, Darlison M. Global epidemiology of haemoglobin disorders and derived service indicators. Bull World Health Organ 2008; 86: 480–487.
    1. Thein SL. The molecular basis of β-thalassemia. Cold Spring Harb Perspect Med 2013; 3: a011700.
    1. Sripichai O, Fucharoen S. Fetal hemoglobin regulation in β-thalassemia: heterogeneity, modifiers and therapeutic approaches. Expert Rev Hematol 2016; 9: 1129–1137.
    1. Liu D, Zhang X, Yu L, et al.. KLF1 mutations are relatively more common in a thalassemia endemic region and ameliorate the severity of β-thalassemia. Blood 2014; 124: 803–811.
    1. Musallam KM, Sankaran VG, Cappellini MD, et al.. Fetal hemoglobin levels and morbidity in untransfused patients with β-thalassemia intermedia. Blood 2012; 119: 364–367.
    1. Forget BG. Molecular basis of hereditary persistence of fetal hemoglobin. Ann N Y Acad Sci 1998; 850: 38–44.
    1. Nuinoon M, Makarasara W, Mushiroda T, et al.. A genome-wide association identified the common genetic variants influence disease severity in β0-thalassemia/hemoglobin E. Hum Genet 2010; 127: 303–314.
    1. Galanello R, Sanna S, Perseu L, et al.. Amelioration of sardinian β0 thalassemia by genetic modifiers. Blood 2009; 114: 3935–3937.
    1. Danjou F, Anni F, Perseu L, et al.. Genetic modifiers of b-thalassemia and clinical severity as assessed by age at first transfusion. Haematologica 2012; 97: 989–993.
    1. Badens C, Joly P, Agouti I, et al.. Variants in genetic modifiers of β-thalassemia can help to predict the major or intermedia type of the disease. Haematologica 2011; 96: 1712–1714.
    1. Uda M, Galanello R, Sanna S, et al.. Genome wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of β-thalassemia. Proc Natl Acad Sci USA 2008; 105: 1620–1625.
    1. Gambari R, Fibach E. Medicinal chemistry of fetal hemoglobin inducers for treatment of β-thalassemia. Curr Med Chem 2007; 14: 199–212.
    1. Lavelle D, Engel JD, Saunthararajah Y. Fetal hemoglobin induction by epigenetic drugs. Semin Hematol 2018; 55: 60–67.
    1. Finotti A, Gambari R. Recent trends for novel options in experimental biological therapy of β-thalassemia. Expert Opin Biol Ther 2014; 14: 1443–1454.
    1. Nuamsee K, Chuprajob T, Pabuprapap W, et al.. Trienone analogs of curcuminoids induce fetal hemoglobin synthesis via demethylation at (G)gamma-globin gene promoter. Sci Rep 2021; 11: 8552.
    1. Antoniani C, Meneghini V, Lattanzi A, et al.. Induction of fetal hemoglobin synthesis by CRISPR/Cas9-mediated editing of the human β-globin locus. Blood 2018; 131: 1960–1973.
    1. Métais JY, Doerfler PA, Mayuranathan T, et al.. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv 2019; 3: 3379–3392.
    1. Mingoia M, Caria CA, Ye L, et al.. Induction of therapeutic levels of HbF in genome-edited primary β(0) 39-thalassaemia haematopoietic stem and progenitor cells. Br J Haematol 2021; 192: 395–404.
    1. Frangoul H, Altshuler D, Cappellini MD, et al.. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia. N Engl J Med 2021; 384: 252–260.
    1. NCT03655678 (A safety and efficacy study evaluating CTX001 in subjects with transfusion-dependent β-thalassemia).
    1. Sehgal SN. Sirolimus: its discovery, biological properties, and mechanism of action. Transplant Proc 2003; 35(3 Suppl): 7S–14S.
    1. Mischiati C, Sereni A, Lampronti I, et al.. Rapamycin-mediated induction of gamma-globin mRNA accumulation in human erythroid cells. Br J Haematol 2004; 126: 612–621.
    1. Fibach E, Bianchi N, Borgatti M, et al.. Effects of rapamycin on accumulation of alpha-, β- and gamma-globin mRNAs in erythroid precursor cells from β-thalassaemia patients. Eur J Haematol 2006; 77: 437–441.
    1. Zuccato C, Bianchi N, Borgatti M, et al.. Everolimus is a potent inducer of erythroid differentiation and gamma-globin gene expression in human erythroid cells. Acta Haematol 2007; 117: 168–176.
    1. Pecoraro A, Troia A, Calzolari R, et al.. Efficacy of rapamycin as inducer of Hb F in primary erythroid cultures from sickle cell disease and β-thalassemia patients. Hemoglobin 2015; 39: 225–229.
    1. Zhang X, Campreciós G, Rimmelé P, et al.. FOXO3-mTOR metabolic cooperation in the regulation of erythroid cell maturation and homeostasis. Am J Hematol 2014; 89: 954–963.
    1. Khaibullina A, Almeida LE, Wang L, et al.. Rapamycin increases fetal hemoglobin and ameliorates the nociception phenotype in sickle cell mice. Blood Cells Mol Dis 2015; 55: 363–372.
    1. Wang J, Tran J, Wang H, et al.. mTOR inhibition improves anaemia and reduces organ damage in a murine model of sickle cell disease. Br J Haematol 2016; 174: 461–469.
    1. Gaudre N, Cougoul P, Bartolucci P, et al.. Improved fetal hemoglobin with mtor inhibitor-based immunosuppression in a kidney transplant recipient with sickle cell disease. Am J Transplant 2017; 17: 2212–2214.
    1. Al-Khatti AA, Alkhunaizi AM. Additive effect of sirolimus and hydroxycarbamide on fetal haemoglobin level in kidney transplant patients with sickle cell disease. Br J Haematol 2019; 185: 959–961.
    1. Ansari SH, Lassi ZS, Khowaja SM, et al.. Hydroxyurea (hydroxycarbamide) for transfusion-dependent β-thalassaemia. Cochrane Database Syst Rev 2019; 3: CD012064.
    1. Algiraigri AH, Wright NAM, Paolucci EO, et al.. Hydroxyurea for lifelong transfusion-dependent β-thalassemia: a meta-analysis. Pediatr Hematol Oncol 2017; 34: 435–448.
    1. Foong WC, Ho JJ, Loh CK, et al.. Hydroxyurea for reducing blood transfusion in non-transfusion dependent β thalassaemias. Cochrane Database Syst Rev 2016; 10: CD011579.
    1. Rigano P, Pecoraro A, Calzolari R, et al.. Desensitization to hydroxycarbamide following long-term treatment of thalassaemia intermedia as observed in vivo and in primary erythroid cultures from treated patients. Br J Haematol 2010; 151: 509–515.
    1. Musallam KM, Rivella S, Vichinsky E, et al.. Non-transfusion-dependent thalassemias. Haematologica 2013; 98: 833–844.
    1. Musallam KM, Rivella S, Taher AT. Management of non-transfusion-dependent beta-thalassemia (NTDT): the next 5 years. Am J Hematol 2021; 96: E57–E59.
    1. Pagani A, Nai A, Silvestri L, et al.. Hepcidin and anemia: a tight relationship. Front Physiol 2019; 10: 1294.
    1. Makis A, Voskaridou E, Papassotiriou I, et al.. Novel therapeutic advances in beta-thalassemia. Biology (Basel) 2021; 10: 546.
    1. NCT03877809 (A personalized medicine approach for β-thalassemia transfusion dependent patients: testing sirolimus in a first pilot clinical trial: SIRTHALACLIN).
    1. Gamberini MR, Prosdocimi M, Gambari R. Sirolimus for treatment of β-thalassemia: from pre-clinical studies to the design of clinical trials. Health Educ Public Health 2021; 4: 425–435.
    1. Musallam KM, Bou-Fakhredin R, Cappellini MD, et al.. 2021 update on clinical trials in beta-thalassemia. Am J Hematol 2021; 96: 1518–1531.
    1. Eldridge SM, Chan CL, Campbell MJ, et al.. CONSORT 2010 statement: extension to randomised pilot and feasibility trials. BMJ 2016; 355: i5239.
    1. Cappellini MD, Farmakis D, Porter J, et al.. Guidelines for the management of transfusion dependent thalassaemia. 4th ed. Nicosia: Thalassemia International Federation, 2021.
    1. Ivanova M, Artusi C, Polo G, et al.. High-throughput LC-MS/MS method for monitoring sirolimus and everolimus in the routine clinical laboratory. Clin Chem Lab Med 2011; 49: 1151–1158.
    1. Navarrete A, Martínez-Alcázar MP, Durán I, et al.. Simultaneous online SPE-HPLC-MS/MS analysis of docetaxel, temsirolimus and sirolimus in whole blood and human plasma. J Chromatogr B Analyt Technol Biomed Life Sci 2013; 921–922: 35–42.
    1. Marinova M, Artusi C, Brugnolo L, et al.. Immunosuppressant therapeutic drug monitoring by LC-MS/MS: workflow optimization through automated processing of whole blood samples. Clin Biochem 2013; 46: 1723–1727.
    1. Wang M, Tang DC, Liu W, et al.. Hydroxyurea exerts bi-modal dose-dependent effects on erythropoiesis in human cultured erythroid cells via distinct pathways. Br J Haematol 2002; 119: 1098–1105.
    1. Fibach E, Bianchi N, Borgatti M, et al.. Mithramycin induces fetal hemoglobin production in normal and thalassemic human erythroid precursor cells. Blood 2003; 102: 1276–1281.
    1. Amoyal I, Goldfarb A, Fibach E. Flow cytometric analysis of hydroxyurea effects on fetal hemoglobin production in cultures of beta-thalassemia erythroid precursors. Hemoglobin 2003; 27: 77–87.
    1. Verschoor CP, Kohli V, Balion C. A comprehensive assessment of immunophenotyping performed in cryopreserved peripheral whole blood. Cytometry B Clin Cytom 2018; 94: 662–670.
    1. Finak G, Langweiler M, Jaimes M, et al.. Standardizing flow cytometry immunophenotyping analysis from the human immunophenotyping consortium. Sci Rep 2016; 6: 20686.
    1. Pinto LA, Trivett MT, Wallace D, et al.. Fixation and cryopreservation of whole blood and isolated mononuclear cells: influence of different procedures on lymphocyte subset analysis by flow cytometry. Cytometry B Clin Cytom 2005; 63: 47–55.
    1. Lechauve C, Keith J, Khandros E, et al.. The autophagy-activating kinase ULK1 mediates clearance of free α-globin in β-thalassemia. Sci Transl Med 2019; 11: eaav4881.
    1. Longo F, Piolatto A, Ferrero GB, et al.. Ineffective erythropoiesis in beta-thalassaemia: key steps and therapeutic options by drugs. Int J Mol Sci 2021; 22: 7229.
    1. Ricchi P, Ammirabile M, Costantini S, et al.. Soluble form of transferrin receptor as a biomarker of overall morbidity in patients with non-transfusion-dependent thalassaemia: a cross-sectional study. Blood Transfus 2016; 14: 538–540.
    1. Khandros E, Kwiatkowski JL. Beta thalassemia: monitoring and new treatment approaches. Hematol Oncol Clin North Am 2019; 33: 339–353.
    1. Bazvand F, Shams S, Borji Esfahani M, et al.. Total antioxidant status in patients with major beta-thalassemia. Iran J Pediatr 2011; 21: 159–165.
    1. Huang Y, Lei Y, Liu R, et al.. Imbalance of erythropoiesis and iron metabolism in patients with thalassemia. Int J Med Sci 2019; 16: 302–310.
    1. Peterson DE, O’Shaughnessy JA, Rugo HS, et al.. Oral mucosal injury caused by mammalian target of rapamycin inhibitors: emerging perspectives on pathobiology and impact on clinical practice. Cancer Med 2016; 5: 1897–1907.
    1. Davies M, Saxena A, Kingswood JC. Management of everolimus-associated adverse events in patients with tuberous sclerosis complex: a practical guide. Orphanet J Rare Dis 2017; 12: 35.
    1. Campistol JM, de Fijter JW, Flechner SM, et al.. mTOR inhibitor-associated dermatologic and mucosal problems. Clin Transplant 2010; 24: 149–156.
    1. Kaplan B, Qazi Y, Wellen JR. Strategies for the management of adverse events associated with mTOR inhibitors. Transplant Rev (Orlando) 2014; 28: 126–133.
    1. Huang J, Nguyen-McCarty M, Hexner EO, et al.. Maintenance of hematopoietic stem cells through regulation of Wnt and mTOR pathways. Nat Med 2012; 18: 1778–1785.
    1. Kahan BD. Sirolimus: a new agent for clinical renal transplantation. Transplant Proc 1997; 29: 48–50.
    1. Vasquez EM. Sirolimus: a new agent for prevention of renal allograft rejection. Am J Health Syst Pharm 2000; 57: 437–448.
    1. Schaffer SA, Ross HJ. Everolimus: efficacy and safety in cardiac transplantation. Expert Opin Drug Saf 2010; 9: 843–854.
    1. Tang CY, Shen A, Wei XF, et al.. Everolimus in de novo liver transplant recipients: a systematic review. Hepatobiliary Pancreat Dis Int 2015; 14: 461–469.
    1. Ji L, Xie W, Zhang Z. Efficacy and safety of sirolimus in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Semin Arthritis Rheum 2020; 50: 1073–1080.
    1. Li H, Ji J, Du Y, et al.. Sirolimus is effective for primary relapsed/refractory autoimmune cytopenia: a multicenter study. Exp Hematol 2020; 89: 87–95.
    1. Wang Q, Luo M, Xiang B, et al.. The efficacy and safety of pharmacological treatments for lymphangioleiomyomatosis. Respir Res 2020; 21: 55.
    1. Sasongko TH, Ismail NF, Zabidi-Hussin Z. Rapamycin and rapalogs for tuberous sclerosis complex. Cochrane Database Syst Rev 2016; 7: CD011272.
    1. Graillon T, Sanson M, Campello C, et al.. Everolimus and octreotide for patients with recurrent meningioma: results from the phase II CEVOREM trial. Clin Cancer Res 2020; 26: 552–557.
    1. Gallo M, Malandrino P, Fanciulli G, et al.. Everolimus as first line therapy for pancreatic neuroendocrine tumours: current knowledge and future perspectives. J Cancer Res Clin Oncol 2017; 143: 1209–1224.
    1. Manohar PM, Beesley LJ, Taylor JM, et al.. Retrospective study of sirolimus and cyclophosphamide in patients with advanced differentiated thyroid cancers. J Thyroid Disord Ther 2015; 4: 188.
    1. Hortobagyi GN. Everolimus plus exemestane for the treatment of advanced breast cancer: a review of subanalyses from BOLERO-2. Neoplasia 2015; 17: 279–288.
    1. Merli M, Ferrario A, Maffioli M, et al.. Everolimus in diffuse large B-cell lymphomas. Future Oncol 2015; 11: 373–383.
    1. Motzer RJ, Escudier B, Oudard S, et al.. Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer 2010; 116: 4256–4265.
    1. Khandros E, Thom CS, D’Souza J, et al.. Integrated protein quality-control pathways regulate free α-globin in murine β-thalassemia. Blood 2012; 119: 5265–5275.
    1. Liu WJ, Ye L, Huang WF, et al.. p62 links the autophagy pathway and the ubiquitin-proteasome system upon ubiquitinated protein degradation. Cell Mol Biol Lett 2016; 21: 29.
    1. Huang Y, Lei Y, Liu R, et al.. Imbalance of erythropoiesis and iron metabolism in patients with thalassemia. Int J Med Sci 2019; 16: 302–310.
    1. Ganz T. Erythropoietic regulators of iron metabolism. Free Radic Biol Med 2019; 133: 69–74.
    1. Lai NS, Yu HC, Huang KY, et al.. Decreased T cell expression of H/ACA box small nucleolar RNA 12 promotes lupus pathogenesis in patients with systemic lupus erythematosus. Lupus 2018; 27: 1499–1508.
    1. Hu S, Wu X, Xu W, et al.. Long-term efficacy and safety of sirolimus therapy in patients with lymphangioleiomyomatosis. Orphanet J Rare Dis 2019; 14: 206.
    1. Wen HY, Wang J, Zhang SX, et al.. Low-dose sirolimus immunoregulation therapy in patients with active rheumatoid arthritis: a 24-week follow-up of the randomized, open-label, parallel-controlled trial. J Immunol Res 2019; 2019: 7684352.
    1. King AJ, Songdej D, Downes DJ, et al.. Reactivation of a developmentally silenced embryonic globin gene. Nat Commun 2021; 12: 4439.
    1. Khamphikham P, Jearawiriyapaisarn N, Tangprasittipap A, et al.. Downregulation of KLF4 activates embryonic and fetal globin mRNA expression in human erythroid progenitor cells. Exp Ther Med 2021; 22: 1105.
    1. Sankaran VG, Xu J, Orkin SH. Transcriptional silencing of fetal hemoglobin by BCL11A. Ann N Y Acad Sci 2010; 1202: 64–68.
    1. Wang X, Angelis N, Thein SL. MYB – a regulatory factor in hematopoiesis. Gene 2018; 665: 6–17.
    1. Ju J, Wang Y, Liu R, et al.. Human fetal globin gene expression is regulated by LYAR. Nucleic Acids Res 2014; 42: 9740–9752.
    1. Yang Y, Ren R, Ly LC, et al.. Structural basis for human ZBTB7A action at the fetal globin promoter. Cell Rep 2021; 36: 109759.
    1. Keating R, Hertz T, Wehenkel M, et al.. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat Immunol 2013; 14: 1266–1276.
    1. Moraschi BF, Noronha IH, Ferreira CP, et al.. Rapamycin improves the response of effector and memory CD8+ T cells induced by immunization with ASP2 of trypanosoma cruzi. Front Cell Infect Microbiol 2021; 11: 676183.

Source: PubMed

3
Abonner