Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Mar 1.
Published in final edited form as: Expert Opin Ther Targets. 2015 Sep 30;20(3):287–301. doi: 10.1517/14728222.2016.1090975

Targeting EPO and EPO RECEPTOR PATHWAYS in Anemia and Dysregulated Erythropoiesis

Nicole Rainville 1, Edward Jachimowicz 1, Don M Wojchowski 1,2,3
PMCID: PMC4829957  NIHMSID: NIHMS773849  PMID: 26419263

Abstract

Introduction

Recombinant human erythropoietin (rhEPO) is a first-line therapeutic for the anemia of chronic kidney disease, cancer chemotherapy, AIDS (Zidovudine therapy) and lower-risk myelodysplastic syndrome. However, rhEPO frequently elevates hypertension, is costly and may affect cancer progression. Potential high merit therefore exists for defining new targets for anti-anemia agents within EPO, and EPO receptor (EPOR) regulatory circuits.

Areas covered

EPO production by renal interstitial fibroblasts is subject to modulation by several regulators of hypoxia-inducible factor 2a (HIF2a) including Iron Response Protein-1, prolyl hydroxylases and HIF2a acetylases, each with potential as anti-anemia drug targets. The cell surface receptor for EPO (EPOR) preassembles as a homodimer (together with JAK2), and novel agents that trigger EPOR complex activation also remain attractive to develop (activating antibodies, mimetics, small molecule agonists). Additionally, certain downstream transducers of EPOR/JAK2 signaling may be drugable including Erythroferrone (a hepcidin regulator), a cytoprotective Spi2a serpin, and select EPOR-associated protein tyrosine phosphatases.

Expert Opinion

While rhEPO (and biosimilars) presently are important mainstay erythropoiesis-stimulating agents, impetus exists for studies of novel ESAs that fortify HIF2a’s effects, act as EPOR agonists, and/or bolster select downstream EPOR pathways to erythroid cell formation. Such agents could lessen rhEPO dosing, side effects and/or costs.

Keywords: erythropoiesis, anemia, erythropoietin (EPO), erythropoiesis stimulating agents (ESAs), EPO receptor, JAK2, hypoxia inducible factors (HIFs), prolyl hydroxylases (PHDs), Spi2a, Erythroferrone, protein tyrosine phosphatases (PTPs), erythroid models

1] EPO, and Present Clinical Uses of rhEPO

Erythropoietin (EPO) is a highly glycosylated sialoglycoprotein hormone1, 2 that is expressed predominantly by (and secreted from) a minor subset of adult renal peritubular interstitial fibroblasts3. During embryonic development, EPO is also produced by precursor neural crest, neural tissue-derived fibroblasts4, and fetal liver cells5. Under conditions of severe anemia, adult hepatic cells additionally can activate EPO gene expression6. EPO levels can be dynamically regulated, with up to 1000-fold increases induced in response to compromised tissue oxygenation7. At perinatal and adult stages, EPO exerts its prime effects on bone marrow resident erythroid progenitor cells (EPCs), and critically supports their survival, growth and development. In canonical pathway contexts, EPO’s cytoprotective effects have been reported to involve the modulation of a subset of (anti)apoptotic factors including Bcl-x, Bcl2, Mcl, and Bim3, 8. Proliferative actions depend upon EPO’s requisite engagement of PI3K/AKT, RAS/MEK/ERK9 and PLCg10 pathways as well as EPOR/JAK2/STAT5 induction of Cyclin D2 and repression of Cyclin G211. Possible effects on erythroid differentiation have been controversial. However, it has recently been demonstrated that EPO has the ability to guide hematopoietic progenitor cells towards erythroid lineage outcomes (at the expense of myelopoiesis)12.

Clinically, recombinant human erythropoietin (rhEPO) and biosimilars13 are used primarily to treat anemia associated with chronic kidney disease (CKD)3, 14, cancer chemotherapy15, Zidovudine therapy among HIV patients 16, blood losses pre- and post surgery17, and low-risk myelodysplastic syndrome (MDS)18, 19. The sustained use of rhEPO over the past three decades as an effective anti-anemia agent points to erythropoietin as one of the most successful recombinant therapeutic agents. Notably, rhEPO can lessen the need for blood transfusions (and inherent risks for alloantibodies, iron overload and acute lung injury)20. In MDS patients, rhEPO also can improve exercise tolerance and quality of life19. For optimal safety, however, rhEPO dosing must be calibrated to achieve hemoglobin levels of 9-12g/dL21-23. As studied among non-dialysis CKD patients, rhEPO dosing to strive for higher hemoglobin levels (13.5g/dL) did not correlate with improved quality of life, and patients in the high hemoglobin arm of this study experienced increased rates of hypertension, myocardial infarction and congestive heart failure22.

One major impetus for pursuing erythropoiesis stimulating agents (ESAs) beyond rhEPO and biosimilars therefore relates to side-effects, with hypertension frequently experienced among CKD patients24-26. While hypertensive effects of rhEPO therapy are well established, underlying mechanisms are not well understood. Endothelin-127-29, and thromboxane30 each has been implicated as rhEPO-modulated vasoconstrictors. Possible contributions of blood cell volume changes or viscosity effects, in contrast, have largely been discounted,25, 26 with transfusions and rhEPO dosing similarly affecting viscosity but not blood pressure. More recently, however, EPOR expression as well as ESA signaling have been reported to be undetectable in primary human endothelial cells31. Further side effects of rhEPO include heightened risks for venous thromboembolism, stroke and death21, 32-35. More controversially, rhEPO has been reported to negatively affect outcomes among patients receiving chemotherapy for certain cancers including breast, head/neck, and additional malignancies15, 36-39. The extent to which tumor cells express EPORs at meaningful densities is also controversial, but EPOR expression levels overall are quite low-level40-42, and with recent notable exceptions40, 41, 43 most EPOR antibodies are not so sensitive or specific40, 42. In studies using an A82 EPOR antibody, EPOR expression in a human cancer cell line panel was nominal44. In xenograft experiments for several human cancer cell lines no significant effects of rhEPO on tumor growth were observed45, and in a study of primary tumor cells from 186 patients, rhEPO did not significantly activate AKT or MAPK signaling46. Nonetheless, in recent studies using an antiserum to a hEPO C-terminal epitope, EPOR expression has been reported among breast cancer cell lines, and associated with estrogen receptor-alpha activation43. Via RNAi approaches, the EPOR furthermore has been indicated to modulate tumor cell growth, survival and/or migration in melanoma47, renal carcinoma48 and breast cancer cell lines43.

Taken together, the above considerations indicate potential high merit for the development of ESAs that safely and effectively stimulate erythropoiesis. This review focuses on possible in-roads to defining new EPO and EPOR targets in the contexts of anemia, and dysregulated erythropoiesis.

2] Targeting HIF2a to heighten endogenous EPO production

When systemic oxygenation is compromised, the expression of EPO within renal interstitial fibroblasts is induced primarily via transcriptional activation3. This depends in part upon interactions of a delineated EPO gene 3’ enhancer with hypoxia inducible factors3, 4, (HIFs, as HIF1a, 2a, 3a coupled to their obligate heterodimeric beta subunit, ARNT)49. Within 5’ EPO gene regulatory domains, roles also have been described for a kidney-inducible region50, and for Gata-2 and -3 repressive effects51. Among three HIFs 1a, 2a, 3a52, Hif2a has been demonstrated via conditional gene deletion studies in mice to be a major Epo gene inducer53. Heightening HIF2a expression therefore provides one potentially attractive in-road to increasing endogenous EPO expression.

During HIF2a transcript translation, major regulatory effects recently have been discovered for Iron Response Protein-1 (IRP1). Specifically, when iron levels are low, apo-IRP1 functions as an RNA binding protein to occupy a unique 5’ stem-loop structure within HIF2a transcripts and hinder translation54. Increased iron levels conversely generate iron-complexed holo-IRP1 which is unable to efficiently bind HIF2a mRNA cis-elements. This reverses suppression and allows for heightened HIF2a translation – together with elevated EPO expression. Unlike Irp2-KO mice which exhibit a microcytic anemia54, 55, Irp1-KO mice develop normally at steady-state. Under modest hypoxia or iron deficiency, Irp1-KO mice, however, exhibit elevated EPO levels and erythroid polycythemia54. In an anti-anemia agent context, this points to apo-IRP1 (within competent renal fibroblasts) as a rational new candidate target whose inhibition is predicted to heighten HIF2a and EPO expression (Figure 1A). DNA and RNA binding proteins (such as IRP1) typically have been considered to be refractive to targeting. Recent reports nonetheless indicate meaningful progress. Examples include initial success via high throughput screens of small molecule inhibitors to the c-Myc RNA binding protein, IMP156; Musashi’s as a family of proto-oncogenic RNA binding proteins57; and to the adipocytic DNA binding protein HMGA258. This elevates prospects for targeting IRP1. Interestingly, IRP1 can also play roles within a cytosolic compartment as an aconitase to catalyze citrate and isocitrate conversions, and enhance NADPH production59. Conditional double knockout studies further indicate roles for Irp1, together with Irp2, in iron adsorption and accumulation in duodenal enterocytes54, 55. In kidney, liver and brown fat, IRP1 levels also are relatively high. The extent to which these properties might complicate IRP1 targeting presently is uncertain.

Figure 1. Approaches to increasing HIF2a levels and/or activity, and heightening endogenous EPO expression.

Figure 1

(A) Upon binding to a unique 5’ domain within HIF2a transcripts, Apo-IRP1 inhibits translation. Small molecule inhibitors that interfere with this interaction therefore have the potential to increase HIF2a and EPO expression together with RBC production. (B) Small molecule inhibitors that inhibit PHD2 have been developed that stabilize HIF2a, and likewise increase EPO expression plus erythrocyte formation. This includes HIF2a inhibitors presently in phase-2 and -3 CKD clinical trials. Such inhibitors also will reinforce PHD2 and HIF2a actions on additional targets, and possible consequences of these latter effects will be important to assess. (C) Acetate supplementation recently has been shown to lessen anemia associated with several forms of stress erythropoiesis. This involves Acss2 mediated Acetyl-CoA production as a limiting step in enhancing HIF2a activity, with EPO as one prime target. Recently, this acetate switch also has been indicated to impact on tumor cell growth and metastasis.

HIF2a additionally is sharply regulated by post-translational modifiers, including Von Hippel-Lindau factor (VHL) and prolyl hydroxylases (PHDs 1-3). VHL is an E3 ubiquitin ligase complex component that was identified among Chuvash polycythemia patients as a loss of function (LOF) mutation60. VHL LOF attenuates the ubiquitination and turnover of HIF2a61, 62. Elevated levels of HIFs then heighten EPO expression, and red cell production. PHDs likewise are negative regulators of HIF’s, and function as prolyl hydroxylases to mark HIF’s for ubiquitination by VHL63. PHDs specifically couple the hydroxylation of prolines within HIFs to the decarboxylation of 2-oxoglutarate (to yield succinate plus CO2)64. Hypoxia inhibits this process, as do an increasing number of pharmacological small molecule inhibitors of PHDs, with most acting as 2-oxoglutarate antagonists64, 65. Biologically, PHDs 1-3 and HIFs 1a-3a are expressed, and exert functional effects within a broad spectrum of tissues that are affected by ischemia65. Pharmacological inhibitors of PHDs therefore are being sought in the contexts of not only anemia but also cardiovascular disease, cerebral ischemia, diabetes, renal disease, retinopathy and cancer metastasis65-67. Via conditional gene disruption studies in mice, PHD2 is emerging as one prime modulator of HIF2a expression in renal EPO producing cells6, 63, 68-72. Recent studies, however, implicate hepatocytes73 as well as osteoblasts74 as meaningful sites of EPO production during hypoxia and anemia. In addition, conditional gene knockout studies are generating insight into relative contributions of PHDs 1-3. Using an Epo gene-GFP mouse model to study renal EPO producing cells (R-EPs) genetic inactivation of PHD2 restored EPO expression, but resulted in polycythemia75. By comparison, compound deletion of PHD1 and PHD3 prevented loss of EPO expression without generating polycythemia. In liver specific conditional knockout experiments, deletion of either PHD1, 2 or 3 was not consequential, but deletion of any two PHDs induced polycythemia73. Compound deletion of PHDs 1, 2 and 3 further generated hepatic fat accumulation. In osteoblasts, effects on EPO production of the compound deletion of PHD1, 2 and -3 were studied74, and relative contributions to HIF stabilization await further investigation.

Which PHDs couple to which HIFs, and under what specific conditions, at-large also are somewhat open questions. PHD2 plus HIF1a are the most evolutionarily conserved couple, the most broadly expressed in mammalian tissues, and a predominant pair under steady-state conditions64. In the regulated expression of EPO and red cell production, roles also have been implicated for a fourth trans-membrane PHD, P4H-TMD76. Conditional gene deletion studies, however, point to PHD2 and HIF2a as major effectors in renal EPO producing cells during stress63, 71, 72. For several HIF2a stabilizing agents, phase 2b and 3 clinical trials are yielding promising initial outcomes in the context of chronic kidney disease associated anemia13. In a partial nephrectomy rat model of chronic kidney disease, one such HIF2a stabilizer also has recently been reported to lessen hypertension as compared to animals dosed with rhEPO (or to partial nephrectomy controls)77. Challenges for HIF stabilizer development and application for anemia include first the desired specificity of PHD inhibitors towards PHDs 1-4 vs. potential advantages of pan-reactivity. This relates to PHD and HIF engagement during stress, including EPO-producing extra-renal tissues. Second, possible consequences of PHD inhibitors on targets other than HIFs, and beyond EPO, merit active consideration. PHDs are involved in IRP2 degradation78, but this effect may help to integrate HIF2a expression with iron levels. PHDs also have been reported to upregulate IKKB79, and to negatively regulate ATF480. PHD inhibitors therefore might heighten NFKB and/or ATF4 activity. Third, HIFs (especially HIF1a and to an extent HIF2a) are known to also regulate angiogenic, glycolytic and certain oncogenesis-associated factors. As recently reviewed by Gilkes and Semenza81, examples of breast cancer targets up-regulated by both HIF1a and 2a include VEGF (Vascular Endothelial Growth Factor), ANGPTL4 (Angiopoietin-Like), CXCR4 (Chemokine C-X-C Motif Receptor), LOX and LOXL4 (Lysyl Oxidase-Like), and Snail1. Based on reported untoward effects of EPO on the progression of select cancers (including breast cancer)15, 36-39, caution might particularly be considered for HIF stabilizer therapy in anemia patients with a history of malignancy.

With further regard to HIF regulation (and targeting) during anemia, heightened attention has also recently been paid to HIF2a’s acetylation. In particular, a requirement during stress erythropoiesis has been identified for Acss2 (acetyl CoA synthetase-2) as a generator of acetyl CoA for this post-translational modification82. In particular, Acss2 gene disruption was unexpectedly observed to worsen anemia due to stress erythropoiesis. In addition, in acquired as well as inherited chronic anemia models, hematocrits were bolstered following acetate supplementation. This proved to involve enhanced HIF2a acetylation and activation by CBP, together with the partnering of CBP with HIF2a. Thus, acetate appears to become metabolically limiting during anemia in ways that affect Hif2a-mediated EPO production (Figure 1C). The extent to which this “acetate switch”, and observed anti-anemia effects of acetate supplementation might translate clinically is presently unknown, but is of significant interest. It is noted, however, that acetate enhancement of HIF signaling may also link stress signaling (and associated pathways) to tumorigenesis as recently demonstrated in a xenograft model for HT1080 fibrosarcoma cell growth and metastasis83.

3] Agonist Activation of EPOR/JAK2 Complexes

During Golgi processing, EPO’s single-pass cell surface receptor (EPOR) preassembles as a homodimer84 (and co-complexes with JAK2)85. Upon ligation, EPOR/JAK2 activation is indicated to occur via trans-membrane conformational signaling as triggered by the bridging of two EPOR chains via EPO site-1 plus site-2 domains86. Site 1 lies within EPO’s D helix and AB coil, and includes N147 and T44 as residues which hydrogen bond to F93 of the EPOR. Site 2 resides within EPO’s A and C helices, including R14 and R10 residues which promote bonding to EPOR M15087. Consistent with this model, mutations at site 1 or 2 residues (e.g., G151A, K45D, N147K) compromise EPO’s binding affinity for the EPOR to <1% of wild-type rhEPO88. Bivalent antibodies with capacities to bind, bridge and activate dimeric EPOR/JAK2 complexes therefore represent a possible in-road to EPOR agonist development. Notably, this agonist approach has been exploited, and in particular for an EPOR activating Ab12.6 antibody and derivatives89, 90. Ab12.6 binds to the EPOR at a delineated epitope distal to rhEPO binding domains, induces a conformational transition, and stimulates JAK2 kinase activity. In a humanized EPOR mouse model (and as administered once monthly at ≥ 0.2mg/kg), AB12.6 increases hematocrits to levels comparable to those stimulated by Darbepoetin-alfa (as injected twice monthly at 0.003 mg/kg). In addition, Ab12.6 variants also have been prepared that exhibit ten-to-thirty-fold attenuated EPOR off-rates (e.g., Ab12.17, Ab12.61, Ab12.76) but unexpectedly are less effective in supporting F36E cell growth and CFUe formation90. Such findings indicate complexities in antibody-induced EPOR/JAK2 activation mechanisms, and/or EPOR trafficking dynamics. For EPOR agonist antibodies, potential advantages include less frequent dosing, lower production cost and a predicted decreased risk for pure red cell aplasia (PRCA). PRCA involves neutralizing anti-EPO antibody production in patients receiving rhEPO, presents as reticulocytopenia plus normocytic anemia91, and is managed using corticosteroids and/or plasmapheresis92. Notably, this receptor activating monoclonal antibody approach has since also been applied to generate a humanized UB22B minibody agonist for the TPO receptor, MPL93. As assessed using primary CD34+ progenitors, UB22B specifically supported megakaryocyte formation, but unlike TPO interestingly did not enhance platelet aggregation93.

A second approach to EPOR agonist development involves the candidate use of small synthetic or recombinant EPO mimetic peptides (EMPs). Proof-of-principle for the notion that small peptide mimetics might be capable of specifically ligating and activating EPOR/JAK2 complexes was first provided via random phage display library based approaches using monomeric peptides (which interestingly proved to lack primary structural homology to rhEPO94). For such single peptide mimetics, however, potencies in vivo were a limiting factor95. More recently, this specific limitation has been overcome through the development of select dimeric EMPs as efficient ESAs. One such approach specifically involved the construction of dimeric recombinant EMPs fused to a humanized Ig Fc scaffold96, 97. Such dimeric mimetibody EMPs exhibit high ESA potency, and sustained erythropoietic activity in mouse and rat models98. As recently reported, this includes mimetibody EMP-induced increases in HbF and HbA as assessed in a mouse model of beta-thalassemia99. Beyond this, and as a notable proof of principle for a fully synthetic mimetic of a complex glycoprotein hormone, PEG-scaffolded dimeric EMPs also have been developed that further proved in initial clinical trials to be non-inferior to rhEPO in treating the anemia of end-stage CKD100. Efficacy in treating PRCA patients also was demonstrated101. Unfortunately, upon expanded clinical use, select CKD patients proved (upon first-time injection) to experience rapid and severe anaphylaxis, leading to discontinuance of this promising ESA construct102. Factors underlying this unexpected off-target effect presently are undetermined, but merit investigation. This includes possible interactions with mast cells with potential involvement of PEG scaffold features (which frequently are drug design components).

A third candidate approach to agonist development relates to certain properties exhibited by EPOR trans- and juxtamembrane regions. Studies of erythroleukemogenic effects of a Friend virus gp55 envelope protein interestingly mapped gp55’s interactions to the EPOR’s transmembrane region103. EPOR mutagenesis studies additionally have defined transmembrane and juxtamembrane regions as activating domains, especially due to cysteine substitutions104. Also, a papillomavirus virus E5 protein that specifically activates PDGF beta-receptor intriguingly has been converted (via random mutagenesis) to a specific EPOR activator that chronically activates the EPOR via transmembrane region targeting105 (Figure 2A). For the latter factor, issues for delivery and potential oncogenic effects are apparent. Nonetheless, and as proof of principle, small molecule therapeutic agents for thrombocytopenia presently are in place (and in practice) that have proven to act by targeting the transmembrane domain of the related homodimeric type-1 receptor for TPO (MPL)106-108 (Figure 2B). In particular, select hydrazine based non-peptide compounds interact with H499 (and likely T496) of MPL’s transmembrane region and stimulate JAK2, downstream targets and thrombopoiesis. In H. sapiens and primates, but not mouse, MPL transmembrane domains contain this histidine residue. This agonist compound also is inactive towards murine Mpl, and in cell lines lacking hMPL106, 109. In addition, related homodimeric receptors for EPO, prolactin, growth hormone and GCSF, a transmembrane residue corresponding to H499 is lacking (Figure 2C), further pointing to apparent specificity of this small molecule agonist. For immune thrombocytopenia, this agonist is indicated to be effective and to exert limited side effects108 and can also act additively with endogenous TPO to increase platelet counts109. In EPOR and anemia contexts, small molecule activators that target the EPOR transmembrane region therefore may provide a rational and relatively untapped in-road to novel ESA development.

Figure 2. Potential targeting of EPOR transmembrane region to activate pre-dimerized EPOR (and JAK2) complexes.

Figure 2

(A) Friend virus envelope protein gp55 has been determined to specifically activate EPOR dimers via interactions with an EPOR transmembrane region. In addition, a 5,000 Mr transmembrane protein has been generated via random mutagenesis of bovine papilloma virus E5 protein (which normally binds and activates PDGFR-beta) that chronically activates the EPOR. (B) As proof-of-principle, small molecule agents that target the transmembrane domain of MPL have been successfully developed as a therapeutic for immune thrombocytopenia, with H499 (and T496) as target sites for one such agent. (C) Comparisons of transmembrane regions (and WSXWS plus box-1) domains within the human homodimeric type-1 receptors for EPO, TPO, prolactin, growth hormone and GCSF.

4] Potential targeting of signal transducers downstream of the EPOR

Certain newly discovered targets and/or pathways that lie downstream of the EPOR, and are intrinsic to erythroid progenitor cells, also may have targeting potential in anemia and/or myeloproliferative disease contexts. Three such examples are considered here. One that might be directly tractable (as a recombinant cytokine) is the C1q /TNF factor, Erythroferrone (ERFE). ERFE was discovered via gene profiling of EPO-response genes in vivo in murine bone marrow and splenic erythroid precursors110. In ERFE-KO mice, EPO or blood loss-induced suppression of hepcidin proved to be defective, thus pointing to a functional role for ERFE in regulating systemic iron levels. ERFE disruption also proved to worsen anemia in a model of B. abortus induced inflammation111. In a th3/+ beta thalassemia intermedia mouse model, ERFE-KO interestingly restored deficit hepcidin levels to normalcy, and moderately decreased hepatic iron overload110. The overall signaling pathway at play here is indicated to involve anemia stimulated ERFE production by (pro)erythroblasts, ERFE suppression of hepcidin production by hepatocytes, and consequently a lessening of hepcidin inhibition of ferroportin-mediated iron efflux from enterocytes, macrophage and hepatocytes112. ERFE therefore has the potential to lessen the anemia of inflammation, and/or to moderate iron overload among thalassemia patients (Figure 3A). Further investigations will be required to assess such effects of ERFE in (pre)clinical models of human anemia and hemoglobinopathies.

Figure 3. Select signal transducers within erythroid precursor cells, and downstream of the EPOR with druggable potential.

Figure 3

(A) One response pathway intrinsic to erythroblasts, and downstream from the EPOR, involves EPO/EPOR/JAK2/Stat5 transcriptional induction of Erythroferrone (ERFE), a C1q-TNF cytokine. As secreted from erythroblasts, ERFE inhibits hepcidin and thereby enhances Ferroportin efflux of systemic iron. ERFE therefore may assist iron deficient anemias, and/or ERFE inhibition may lessen iron overload as associated, for example, with thalassemia. (B) Via studies of an Spi2A Serpin (as an additional novel EPO/EPOR/JAK2/Stat5 target gene) erythroblast lysosomal membrane permeability has been shown to become compromised during stress erythropoiesis, especially in oxidative and elevated ROS contexts. Small molecule inhibitors of leached lysosomal executioner cathepsins B (and L) therefore represent rational new targets as anti-anemia agents and erythroblast cytoprotectants during such stress erythropoiesis (e.g., sickle cell anemia, thalassemia). (C) Among protein tyrosine phosphatases engaged in erythroid precursors via the EPOR, PTPN6 as a negative effector is a potential target in anti-anemia contexts. As a positive effector, PTPN18 might be considered as a new target in a context of lessening EPOR/JAK2 activation or effects for polycythemia and/or myeloproliferative disease.

During late-stage development, erythroblasts accumulate high-level iron, initiate heme plus hemoglobin biosynthesis, and during this process generate a strongly oxidizing milieu113. This further involves increased production of ROS due to pressure on oxidative phosphorylation114, with iron also acting as a potent catalyst for ROS-mediated oxidative damage115. Attention to this aspect of erythroblast biology has led to the recent discovery of a Serpina3g/Spi2A intracellular serpin as an unexpected EPOR/JAK2/Stat5 induced gene whose product exerts important cytoprotective roles during stress erythropoiesis116. In particular, Serpina3g/Spi2A gene disruption results in substantial losses of late stage erythroblasts in the wake of oxidant induced hemolysis, or sub-lethal irradiation. Mechanistically, this proved to involve Spi2A inhibition of an excecutioner Cathepsin B as leached from compromised erythroblast lysosomes. In addition, Spi2A’s effects were phenocopied by a small molecule inhibitor of Cathepsin B. Leached lysosomal cathepsins within erythroblasts that form during stress erythropoiesis therefore represent a new candidate target for at least certain forms of dysregulated erythropoiesis116 (Figure 3B). In thalassemia and sickle cell disease, for example, erythroid precursors can accelerate the production of oxidants113 which may then compromise lysosomes and lead to executioner cathepsin release. In oncology contexts, Cathepsin B also is emerging as a druggable target, and biochemically Cathepsins are a tractable target for high-throughput screening117.

EPO ligation of its receptor (plus JAK2 activation) is also known to lead to the engagement of select protein tyrosine kinases (PTPs). In early studies, these specifically were defined as the SH2 domain-encoding PTPs PTPN11 / SHP2118, and PTPN6 / SHP1119. In addition, a non-SH2 PTP, PTPN18, has recently been reported as a novel EPO/EPOR/JAK2 signal transducing factor120. PTPN11 is becoming well studied, and this in part is due to the occurrence of dysregulating PTPN11 mutations associated with myeloproliferative disease, juvenile myelomonocytic leukemia, and within a neural context, Noonan syndrome121. During erythropoiesis, mice harboring a PTPN11 D61Y mutation have been observed to hyper-expand early-stage pro-erythroblasts, with heightened Stat3, Akt and Erk activation also exhibited122. In studies by Sharma et al123, 124, PTPN11 also has been shown to be a mediator of oncogenic KIT effects on mastocytosis. PTPN11 therefore acts predominantly as an overall positive effector within select type 1 cytokine receptor and RTK systems. PTPN6, in contrast, is expressed primarily in hematopoietic cells, and acts predominantly as a negative regulator119, 125. As examples, the LOF mutation of PTPN6 in “motheaten” mice126 or the mutation of EPOR PY motifs for PTPN6 docking127 each result in hyper-responsiveness of erythroid precursors to EPO, and heightened JAK2 activation. PTPN18 is less studied, is known to be associated with HER2 signaling128, and recently has been described as a rapidly pY-phosphorylated EPO/EPOR signaling component and positively acting regulator of erythroid precursor cell growth120. Unlike PTPN6 and PTPN11, PTPN18 is less complex in its structure, lacks SH2 co-factor partnering domains, and occurs predominantly as a cytosolic tyrosine phosphatase128. In human erythroid cells PTPN18 also exhibits stage-selective expression, with multi-fold elevated expression at a CFUe stage (see NCBI GEO GDS3860).

In EPOR and anemia contexts, targeting PTPN11 is counter-intuitive due to its predominant roles as a positive effector118, 119, 121, 122, 124. PTPN11 inhibitors, however, are successfully being developed for candidate use in juvenile myelomonocytic leukemia and Noonan disease129-131. For such inhibitors attention should be paid to possible compromising effects on erythropoiesis. As a negatively acting EPOR effector, PTPN6 might be attractive as a target in select anemia contexts. Certain risks, however, may exist for inhibitor-induced hyperproliferation of myeloid and/or lymphoid hematopoietic cell populations126. Finally, based on emerging roles for PTPN18 as a positive effector of EPO-dependent erythropoiesis120, inhibitors to PTPN18 while predicted to attenuate erythropoiesis, nonetheless might prove to be valuable in erythropolycythemia and certain myeloproliferative neoplasm contexts (Figure 3C). This potentially includes JAK2 V617F associated myeloproliferative disease, for which new in-roads beyond (or in addition to) existing JAK2 inhibitors is emerging132. Until recently, PTPs have been considered to be refractive targets for small molecule inhibitors130. Complications involve a deep active site that is positively charged, readily attracts negatively charged compounds, and is susceptible to oxidation (limiting, for example, the use of redox cycling compounds screens). Challenges for specificity also exist due to the large overall number of PTPs130. For select PTPs, progress towards specific small molecule inhibition nonetheless has been made as recently summarized for PTPN11131, PTP1B133 and lymphoid-specific tyrosine phosphatase134. In part, this has involved the defining of co-targetable surfaces and pockets that lie adjacent to PTP catalytic domains, together with the development of select bidentate inhibitors135.

5] Model systems for the discovery and validation of new druggable EPO/EPOR/JAK2 targets and pathways

Among lower vertebrates (e.g., Zebrafish, Xenopus), EPORs are represented, but are not reactive with rhEPO, and lack several functional cytoplasmic phosphotyrosine (pY) signaling motifs that have evolved within murine and human EPORs. The murine EPOR is efficiently activated by rhEPO, and has 86% identity with the hEPO including eight of nine pY signaling motifs. For mouse models, genetic approaches in particular are time intensive, but nonetheless have been advantageous via the development, for example, of useful models for the anemia of chronic renal disease110, chemotherapy98, sublethal irradiation136, blood loss110 and hemoglobinopathies99. In addition, mEPOR-KO mice expressing the hEPOR have been generated and employed in studies of EPOR agonists89. For the challenge of discovering and characterizing new EPO and EPOR targets, value for such murine models is illustrated via studies of HIF2a as a prime EPO inducer6, 63; PHD2 as a central inhibitor of HIF2a13; Erythroferrone as a novel hepcidin suppressor110, 111; the discovery of lysosomal compromise and cathepsin leaching during stress erythropoiesis116; and the emerging utility of a GDF11-adsorbing soluble Activin IIA and IIB receptors as anti-anemia agents137.

In studies of anemia and stress erythropoiesis, murine models nonetheless can present certain potential complications. In mouse, the spleen can rapidly become engaged as a major erythroid organ to expand the erythron138, 139, and this may involve Hedgehog and BMP4 cytokine effects that may not be as critical for human stress erythropoiesis140, 141. In addition, recent comparisons of murine and human expressed transcriptomes in parallel EPC developmental series reveal several unexpected major differences142, 143. As examples, the induction of late-stage factors in murine EPCs is more accelerated; MAPK signaling modules are incongruous in their up- vs down-modulated components142; and non-coding RNA populations are particularly disparate including several that promote the late-stage development of murine EPCs but are absent from H. sapiens143. Differences between mouse and human erythropoiesis also are illustrated (for example) by the lack of disease phenotypes in mice harboring a hematopoietic deficiency in SEC23B144 (a condition genetically known to underlie congenital dyserythropoietic anemia type II), and by the discovery of C1ORF186/”RHEX” as a novel EPO/EPOR target and molecular adaptor which has evolved in H sapiens EPCs but not in mice, rats or lower vertebrates145.

For EPO/EPOR targeting concerns, the above considerations point to advantages in employing human erythroid precursors. For human EPCs, major advances have been made for two-phase serum-free ex vivo systems that employ dexamethasone to efficiently generate CFUe and (pro)erythroblasts145, and for three-phase systems that further support development to anucleated erythrocytes142. As primary sources, peripheral blood monocytes, mobilized CD34pos progenitors, or cord blood progenitors can be used. Recent progress also has been made in the use of iPS cells. Like hES cells, one complication for iPS EPCs involves the expression of ontogenically early fetal and embryonic HB-F and HB-E rather than adult hemoglobin (HB-A). For EPOR targeting studies, however, this limitation may not be so compromising, and can be overcome via ectopic co-expression of the adult globin activators KLF1 plus Bcl11A-XL146. Progress is also being made towards the establishment of standardized, feeder layer-free conditions for erythroid differentiation. In addition, in recent studies of iPS cell formation from cord blood, a system has been defined that yields up to 1068 expansion of (pro)erythroblasts that retain their capacity to undergo late-stage differentiation147. Finally, for EPO and EPOR-related myeloproliferative neoplasms, including JAK2-V617F associated disease, patient-derived iPS cells illustrate the potential high value for drug targeting studies. As one illustrating example, iPS cells that are hemizygous for JAK2-V617F develop as megakaryocytes to HGF-independent blasts, but transformation to EPO-independence unexpectedly does not occur within the erythroid lineage148. Another clinically relevant opportunity for erythroid iPS cell investigations is given by Calcineurin mutations which also have recently been linked to MPN, and Stat5 signaling149.

6] SUMMARY, AND OPINION

Presently, rhEPO and biosimilars continue to serve as important mainstay therapeutics for anemia associated with CDK, cancer chemotherapy, lower-risk MDS, and AIDS Zidovudine regimens3, 14-19. Outcomes can include lessening of transfusions, together with gains in hemoglobin levels, tissue oxygenation, and quality of life. However, intravenous dosing can be frequent, side-effects can be compromising, and costs to health systems can be high (see Introduction). Targets within EPO-producing cells that bolster endogenous EPO expression therefore first continue to be attractive. This includes EPO producing renal interstitial fibroblasts, as well as hepatocytes and osteoblasts each of which has been demonstrated to activate EPO expression upon PHD inactivation73, 74. Promise, and potential complications of PHD inhibitor approaches to treating anemia are summarized in Section 2. For the anemia of chronic kidney disease, an additional potential complication relates to renal fibrosis. Studies involving genetic or pharmacological inactivation of PHDs have demonstrated that EPO expression in EPO-low cells (which can convert to myelofibroblasts) can be reactivated75, 150. The extent to which this might be durably sustained (or perhaps progressively diminished) in patients with end-stage renal disease becomes one important question to address. Possible activation of EPO expression in extra-renal tissues (liver, osteoblasts) therefore may also become important to assess. As an RNA binding protein, IRP1 will be a challenging target. Apo-IRP1 inhibitors also would need to be innocuous towards IRP2, which regulates several key iron binding proteins55. Apo-IRP1 inhibition nonetheless would circumvent possible side-effects of PHD inhibition. For example, genetic inactivation of PHD2 and PHD3 sensitizes mice to beta-adrenergic stress induced myocardial injury151, while genetic or pharmacological inhibition of PHD2 limits functional recovery post corticospinal tract injury to the sensorimotor cortex152.

Promise also continues to exist for the development of ESAs that efficiently activate preassembled EPOR dimers89, 90, 96-99. This prospectively includes scaffolded dimeric EPO mimetic peptides that bind EPORs at EPO binding sites as well as small molecule dimerizing agents that might act additively with rhEPO. Exciting progress furthermore continues to be made in discovering new functionally important EPO/EPOR signal transducers and response pathways that may become druggable. Illustrating examples include Erythroferrone as an EPO-induced hepcidin suppressor110, 111, and ROS-induced lysosomal leaching of executioner cathepsins within stressed erythroblasts116. Success in targeting EPO/EPOR signal transduction factors intrinsic to erythroid precursor cells also has the potential advantage of lessening EPO/ESA dosing, and side-effects. Ongoing and future transcriptome and proteomic studies are also likely to reveal novel pro-erythropoietic factors (such as PTPN18)120 as rational new targets for inhibition in the context of myeloproliferative neoplasms. Importantly, continued major advancements in the ability to propagate and differentiate primary human erythroid progenitor cells provide enriched opportunities for target interrogations, drug screening and validations146-148.

ACKNOWLEDGMENTS

Support for this work was provided by NIH R01 HL044491 (DMW, PI). Additionally, the authors thank Karen Miller for substantial and sustained administrative contributions to manuscript preparation.

Footnotes

AUTHOR CONTRIBUTIONS: All authors contributed in substantial ways to subject conceptualization, literature research and manuscript construction. This includes prime contributions by NR to sections 1 and 3, and by EJ to section 5.

LITERATURE CITED

  • 1.Kawasaki N, Ohta M, Hyuga S, Hyuga M, Hayakawa T. Application of liquid chromatography/mass spectrometry and liquid chromatography with tandem mass spectrometry to the analysis of the site-specific carbohydrate heterogeneity in erythropoietin. Analytical biochemistry. 2000 Oct 1;285(1):82–91. doi: 10.1006/abio.2000.4739. [DOI] [PubMed] [Google Scholar]
  • 2.Froehlich JW, Barboza M, Chu C, Lerno LA, Jr., Clowers BH, Zivkovic AM, et al. Nano-LC-MS/MS of glycopeptides produced by nonspecific proteolysis enables rapid and extensive site-specific glycosylation determination. Analytical chemistry. 2011 Jul 15;83(14):5541–7. doi: 10.1021/ac2003888. [DOI] [PubMed] [Google Scholar]
  • 3.Bunn HF. Erythropoietin. Cold Spring Harbor perspectives in medicine. 2013 Mar;3(3):a011619. doi: 10.1101/cshperspect.a011619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Suzuki N, Hirano I, Pan X, Minegishi N, Yamamoto M. Erythropoietin production in neuroepithelial and neural crest cells during primitive erythropoiesis. Nature communications. 2013;4:2902. doi: 10.1038/ncomms3902. [DOI] [PubMed] [Google Scholar]
  • 5.Koury ST, Bondurant MC, Koury MJ. Localization of erythropoietin synthesizing cells in murine kidneys by in situ hybridization. Blood. 1988 Feb;71(2):524–7. [PubMed] [Google Scholar]
  • 6.Minamishima YA, Kaelin WG., Jr. Reactivation of hepatic EPO synthesis in mice after PHD loss. Science. 2010 Jul 23;329(5990):407. doi: 10.1126/science.1192811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Becker V, Schilling M, Bachmann J, Baumann U, Raue A, Maiwald T, et al. Covering a broad dynamic range: information processing at the erythropoietin receptor. Science. 2010 Jun 11;328(5984):1404–8. doi: 10.1126/science.1184913. [DOI] [PubMed] [Google Scholar]
  • 8.Kuhrt D, Wojchowski DM. Emerging EPO and EPO receptor regulators, and signal transducers. Blood. 2015 Apr 17; doi: 10.1182/blood-2014-11-575357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Arcasoy MO, Jiang X. Co-operative signalling mechanisms required for erythroid precursor expansion in response to erythropoietin and stem cell factor. British journal of haematology. 2005 Jul;130(1):121–9. doi: 10.1111/j.1365-2141.2005.05580.x. [DOI] [PubMed] [Google Scholar]
  • 10.Schnoder TM, Arreba-Tutusaus P, Griehl I, Bullinger L, Buschbeck M, Lane SW, et al. Epo-induced erythroid maturation is dependent on Plcgamma1 signaling. Cell death and differentiation. 2015 Jun;22(6):974–85. doi: 10.1038/cdd.2014.186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Fang J, Menon M, Kapelle W, Bogacheva O, Bogachev O, Houde E, et al. EPO modulation of cell-cycle regulatory genes, and cell division, in primary bone marrow erythroblasts. Blood. 2007 Oct 1;110(7):2361–70. doi: 10.1182/blood-2006-12-063503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Grover A, Mancini E, Moore S, Mead AJ, Atkinson D, Rasmussen KD, et al. Erythropoietin guides multipotent hematopoietic progenitor cells toward an erythroid fate. The Journal of experimental medicine. 2014 Feb 10;211(2):181–8. doi: 10.1084/jem.20131189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Jelkmann W. The ESA scenario gets complex: from biosimilar epoetins to activin traps. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2015 Apr;30(4):553–59. doi: 10.1093/ndt/gfu089. [DOI] [PubMed] [Google Scholar]
  • 14.Ramanath V, Gupta D, Jain J, Chaudhary K, Nistala R. Anemia and chronic kidney disease: making sense of the recent trials. Reviews on recent clinical trials. 2012 Aug;7(3):187–96. doi: 10.2174/157488712802281222. [DOI] [PubMed] [Google Scholar]
  • 15.Oster HS, Neumann D, Hoffman M, Mittelman M. Erythropoietin: the swinging pendulum. Leukemia research. 2012 Aug;36(8):939–44. doi: 10.1016/j.leukres.2012.04.017. [DOI] [PubMed] [Google Scholar]
  • 16.Sullivan PS, Hanson DL, Richardson JT, Brooks JT. Trends in the Treatment of Anemia Using Recombinant Human Erythropoietin in Patients with HIV Infection. The open AIDS journal. 2011;5:113–8. doi: 10.2174/1874613601105010113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Tran DH, Wong GT, Chee YE, Irwin MG. Effectiveness and safety of erythropoiesis-stimulating agent use in the perioperative period. Expert opinion on biological therapy. 2014 Jan;14(1):51–61. doi: 10.1517/14712598.2014.858116. [DOI] [PubMed] [Google Scholar]
  • 18.Komrokji RS, Lancet JE, Swern AS, Chen N, Paleveda J, Lush R, et al. Combined treatment with lenalidomide and epoetin alfa in lower-risk patients with myelodysplastic syndrome. Blood. 2012 Oct 25;120(17):3419–24. doi: 10.1182/blood-2012-03-415661. [DOI] [PubMed] [Google Scholar]
  • 19.Hellstrom-Lindberg E, van de Loosdrecht A. Erythropoiesis stimulating agents and other growth factors in low-risk MDS. Best practice & research Clinical haematology. 2013 Dec;26(4):401–10. doi: 10.1016/j.beha.2013.09.007. [DOI] [PubMed] [Google Scholar]
  • 20.Tanhehco YC, Berns JS. Red blood cell transfusion risks in patients with end-stage renal disease. Seminars in dialysis. 2012 Sep-Oct;25(5):539–44. doi: 10.1111/j.1525-139X.2012.01089.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rizzo JD, Brouwers M, Hurley P, Seidenfeld J, Arcasoy MO, Spivak JL, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood. 2010 Nov 18;116(20):4045–59. doi: 10.1182/blood-2010-08-300541. [DOI] [PubMed] [Google Scholar]
  • 22.Singh AK, Szczech L, Tang KL, Barnhart H, Sapp S, Wolfson M, et al. Correction of anemia with epoetin alfa in chronic kidney disease. The New England journal of medicine. 2006 Nov 16;355(20):2085–98. doi: 10.1056/NEJMoa065485. [DOI] [PubMed] [Google Scholar]
  • 23.Horl WH. Anaemia management and mortality risk in chronic kidney disease. Nature reviews Nephrology. 2013 May;9(5):291–301. doi: 10.1038/nrneph.2013.21. [DOI] [PubMed] [Google Scholar]
  • 24.Strippoli GF, Craig JC, Manno C, Schena FP. Hemoglobin targets for the anemia of chronic kidney disease: a meta-analysis of randomized, controlled trials. Journal of the American Society of Nephrology : JASN. 2004 Dec;15(12):3154–65. doi: 10.1097/01.ASN.0000145436.09176.A7. [DOI] [PubMed] [Google Scholar]
  • 25.Krapf R, Hulter HN. Arterial hypertension induced by erythropoietin and erythropoiesis-stimulating agents (ESA) Clinical journal of the American Society of Nephrology : CJASN. 2009 Feb;4(2):470–80. doi: 10.2215/CJN.05040908. [DOI] [PubMed] [Google Scholar]
  • 26.Vaziri ND, Zhou XJ. Potential mechanisms of adverse outcomes in trials of anemia correction with erythropoietin in chronic kidney disease. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 2009 Apr;24(4):1082–8. doi: 10.1093/ndt/gfn601. [DOI] [PubMed] [Google Scholar]
  • 27.Kanbay M, Akcay A, Delibasi T, Uz B, Kaya A, Koca C, et al. Comparison of effects of darbepoetin alfa and epoetin alfa on serum endothelin level and blood pressure. Advances in therapy. 2007 Mar-Apr;24(2):346–52. doi: 10.1007/BF02849903. [DOI] [PubMed] [Google Scholar]
  • 28.Barhoumi T, Briet M, Kasal DA, Fraulob-Aquino JC, Idris-Khodja N, Laurant P, et al. Erythropoietin-induced hypertension and vascular injury in mice overexpressing human endothelin-1: exercise attenuated hypertension, oxidative stress, inflammation and immune response. Journal of hypertension. 2014 Apr;32(4):784–94. doi: 10.1097/HJH.0000000000000101. [DOI] [PubMed] [Google Scholar]
  • 29.Briet M, Barhoumi T, Mian MO, Sierra C, Boutouyrie P, Davidman M, et al. Effects of recombinant human erythropoietin on resistance artery endothelial function in stage 4 chronic kidney disease. Journal of the American Heart Association. 2013 Apr;2(2):e000128. doi: 10.1161/JAHA.113.000128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Heinisch BB, Vcelar B, Kapiotis S, Blann A, Wolzt M, Siller-Matula JM, et al. The effect of erythropoietin on platelet and endothelial activation markers: a prospective trial in healthy volunteers. Platelets. 2012;23(5):352–8. doi: 10.3109/09537104.2011.631621. [DOI] [PubMed] [Google Scholar]
  • 31.Sinclair AM, Coxon A, McCaffery I, Kaufman S, Paweletz K, Liu L, et al. Functional erythropoietin receptor is undetectable in endothelial, cardiac, neuronal, and renal cells. Blood. 2010 May 27;115(21):4264–72. doi: 10.1182/blood-2009-10-248666. [DOI] [PubMed] [Google Scholar]
  • 32.Glaspy J. Update on safety of ESAs in cancer-induced anemia. Journal of the National Comprehensive Cancer Network : JNCCN. 2012 May;10(5):659–66. doi: 10.6004/jnccn.2012.0065. [DOI] [PubMed] [Google Scholar]
  • 33.Glaspy J, Crawford J, Vansteenkiste J, Henry D, Rao S, Bowers P, et al. Erythropoiesis-stimulating agents in oncology: a study-level meta-analysis of survival and other safety outcomes. British journal of cancer. 2010 Jan 19;102(2):301–15. doi: 10.1038/sj.bjc.6605498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pfeffer MA, Burdmann EA, Chen CY, Cooper ME, de Zeeuw D, Eckardt KU, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. The New England journal of medicine. 2009 Nov 19;361(21):2019–32. doi: 10.1056/NEJMoa0907845. [DOI] [PubMed] [Google Scholar]
  • 35.Bennett CL, Silver SM, Djulbegovic B, Samaras AT, Blau CA, Gleason KJ, et al. Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. Jama. 2008 Feb 27;299(8):914–24. doi: 10.1001/jama.299.8.914. [DOI] [PubMed] [Google Scholar]
  • 36.Leyland-Jones B, Semiglazov V, Pawlicki M, Pienkowski T, Tjulandin S, Manikhas G, et al. Maintaining normal hemoglobin levels with epoetin alfa in mainly nonanemic patients with metastatic breast cancer receiving first-line chemotherapy: a survival study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2005 Sep 1;23(25):5960–72. doi: 10.1200/JCO.2005.06.150. [DOI] [PubMed] [Google Scholar]
  • 37.Henke M, Laszig R, Rube C, Schafer U, Haase KD, Schilcher B, et al. Erythropoietin to treat head and neck cancer patients with anaemia undergoing radiotherapy: randomised, double-blind, placebo-controlled trial. Lancet. 2003 Oct 18;362(9392):1255–60. doi: 10.1016/S0140-6736(03)14567-9. [DOI] [PubMed] [Google Scholar]
  • 38.Untch M, von Minckwitz G, Konecny GE, Conrad U, Fett W, Kurzeder C, et al. PREPARE trial: a randomized phase III trial comparing preoperative, dose-dense, dose-intensified chemotherapy with epirubicin, paclitaxel, and CMF versus a standard-dosed epirubicin-cyclophosphamide followed by paclitaxel with or without darbepoetin alfa in primary breast cancer--outcome on prognosis. Annals of oncology : official journal of the European Society for Medical Oncology / ESMO. 2011 Sep;22(9):1999–2006. doi: 10.1093/annonc/mdq713. [DOI] [PubMed] [Google Scholar]
  • 39.Debeljak N, Solar P, Sytkowski AJ. Erythropoietin and cancer: the unintended consequences of anemia correction. Frontiers in immunology. 2014;5:563. doi: 10.3389/fimmu.2014.00563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Elliott S, Busse L, McCaffery I, Rossi J, Sinclair A, Spahr C, et al. Identification of a sensitive anti-erythropoietin receptor monoclonal antibody allows detection of low levels of EpoR in cells. Journal of immunological methods. 2010 Jan 31;352(1-2):126–39. doi: 10.1016/j.jim.2009.10.006. [DOI] [PubMed] [Google Scholar]
  • 41.Singh S, Verma R, Pradeep A, Leu K, Mortensen RB, Young PR, et al. Dynamic ligand modulation of EPO receptor pools, and dysregulation by polycythemia-associated EPOR alleles. PloS one. 2012;7(1):e29064. doi: 10.1371/journal.pone.0029064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Elliott S, Busse L, Bass MB, Lu H, Sarosi I, Sinclair AM, et al. Anti-Epo receptor antibodies do not predict Epo receptor expression. Blood. 2006 Mar 1;107(5):1892–5. doi: 10.1182/blood-2005-10-4066. [DOI] [PubMed] [Google Scholar]
  • 43.Reinbothe S, Larsson AM, Vaapil M, Wigerup C, Sun J, Jogi A, et al. EPO-independent functional EPO receptor in breast cancer enhances estrogen receptor activity and promotes cell proliferation. Biochemical and biophysical research communications. 2014 Feb 28;445(1):163–9. doi: 10.1016/j.bbrc.2014.01.165. [DOI] [PubMed] [Google Scholar]
  • 44.Swift S, Ellison AR, Kassner P, McCaffery I, Rossi J, Sinclair AM, et al. Absence of functional EpoR expression in human tumor cell lines. Blood. 2010 May 27;115(21):4254–63. doi: 10.1182/blood-2009-10-248674. [DOI] [PubMed] [Google Scholar]
  • 45.Kataoka M, Moriya Y, Moriguchi Y, Iwai T, Fujimoto-Ouchi K, Shirane M, et al. Effect of erythropoietin on human tumor growth in xenograft models. Molecular medicine reports. 2010 Jan-Feb;3(1):95–101. doi: 10.3892/mmr_00000224. [DOI] [PubMed] [Google Scholar]
  • 46.Patterson SD, Rossi JM, Paweletz KL, Fitzpatrick VD, Begley CG, Busse L, et al. Functional EpoR pathway utilization is not detected in primary tumor cells isolated from human breast, non-small cell lung, colorectal, and ovarian tumor tissues. PloS one. 2015;10(3):e0122149. doi: 10.1371/journal.pone.0122149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Kumar SM, Zhang G, Bastian BC, Arcasoy MO, Karande P, Pushparajan A, et al. Erythropoietin receptor contributes to melanoma cell survival in vivo. Oncogene. 2012 Mar 29;31(13):1649–60. doi: 10.1038/onc.2011.366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wu P, Zhang N, Wang X, Zhang C, Li T, Ning X, et al. The erythropoietin/erythropoietin receptor signaling pathway promotes growth and invasion abilities in human renal carcinoma cells. PloS one. 2012;7(9):e45122. doi: 10.1371/journal.pone.0045122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Labrecque MP, Prefontaine GG, Beischlag TV. The aryl hydrocarbon receptor nuclear translocator (ARNT) family of proteins: transcriptional modifiers with multi-functional protein interfaces. Current molecular medicine. 2013 Aug;13(7):1047–65. doi: 10.2174/15665240113139990042. [DOI] [PubMed] [Google Scholar]
  • 50.Madan A, Lin C, Hatch SL, 2nd, Curtin PT. Regulated basal, inducible, and tissue-specific human erythropoietin gene expression in transgenic mice requires multiple cis DNA sequences. Blood. 1995 May 15;85(10):2735–41. [PubMed] [Google Scholar]
  • 51.Obara N, Suzuki N, Kim K, Nagasawa T, Imagawa S, Yamamoto M. Repression via the GATA box is essential for tissue-specific erythropoietin gene expression. Blood. 2008 May 15;111(10):5223–32. doi: 10.1182/blood-2007-10-115857. [DOI] [PubMed] [Google Scholar]
  • 52.Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity. 2014 Oct 16;41(4):518–28. doi: 10.1016/j.immuni.2014.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Haase VH. Regulation of erythropoiesis by hypoxia-inducible factors. Blood reviews. 2013 Jan;27(1):41–53. doi: 10.1016/j.blre.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Anderson SA, Nizzi CP, Chang YI, Deck KM, Schmidt PJ, Galy B, et al. The IRP1-HIF-2alpha axis coordinates iron and oxygen sensing with erythropoiesis and iron absorption. Cell metabolism. 2013 Feb 5;17(2):282–90. doi: 10.1016/j.cmet.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wilkinson N, Pantopoulos K. The IRP/IRE system in vivo: insights from mouse models. Frontiers in pharmacology. 2014;5:176. doi: 10.3389/fphar.2014.00176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Mahapatra L, Mao C, Andruska N, Zhang C, Shapiro DJ. High-throughput fluorescence anisotropy screen for inhibitors of the oncogenic mRNA binding protein, IMP-1. Journal of biomolecular screening. 2014 Mar;19(3):427–36. doi: 10.1177/1087057113499633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wu X, Lan L, Wilson DM, Marquez RT, Tsao WC, Gao P, et al. Identification and Validation of Novel Small Molecule Disruptors of HuR-mRNA Interaction. ACS chemical biology. 2015 Mar;:17. doi: 10.1021/cb500851u. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Alonso N, Guillen R, Chambers JW, Leng F. A rapid and sensitive high-throughput screening method to identify compounds targeting protein-nucleic acids interactions. Nucleic acids research. 2015 Apr 30;43(8):e52. doi: 10.1093/nar/gkv069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ferecatu I, Goncalves S, Golinelli-Cohen MP, Clemancey M, Martelli A, Riquier S, et al. The diabetes drug target MitoNEET governs a novel trafficking pathway to rebuild an Fe-S cluster into cytosolic aconitase/iron regulatory protein 1. The Journal of biological chemistry. 2014 Oct 10;289(41):28070–86. doi: 10.1074/jbc.M114.548438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Russell RC, Sufan RI, Zhou B, Heir P, Bunda S, Sybingco SS, et al. Loss of JAK2 regulation via a heterodimeric VHL-SOCS1 E3 ubiquitin ligase underlies Chuvash polycythemia. Nature medicine. 2011 Jul;17(7):845–53. doi: 10.1038/nm.2370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ang SO, Chen H, Hirota K, Gordeuk VR, Jelinek J, Guan Y, et al. Disruption of oxygen homeostasis underlies congenital Chuvash polycythemia. Nature genetics. 2002 Dec;32(4):614–21. doi: 10.1038/ng1019. [DOI] [PubMed] [Google Scholar]
  • 62.Bader HL, Hsu T. Systemic VHL gene functions and the VHL disease. FEBS letters. 2012 Jun 4;586(11):1562–9. doi: 10.1016/j.febslet.2012.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Franke K, Gassmann M, Wielockx B. Erythrocytosis: the HIF pathway in control. Blood. 2013 Aug 15;122(7):1122–8. doi: 10.1182/blood-2013-01-478065. [DOI] [PubMed] [Google Scholar]
  • 64.Bishop T, Ratcliffe PJ. HIF Hydroxylase Pathways in Cardiovascular Physiology and Medicine. Circulation research. 2015 Jun 19;117(1):65–79. doi: 10.1161/CIRCRESAHA.117.305109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Selvaraju V, Parinandi NL, Adluri RS, Goldman JW, Hussain N, Sanchez JA, et al. Molecular mechanisms of action and therapeutic uses of pharmacological inhibitors of HIF-prolyl 4-hydroxylases for treatment of ischemic diseases. Antioxidants & redox signaling. 2014 Jun 1;20(16):2631–65. doi: 10.1089/ars.2013.5186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Goldmacher VS, Lambert JM, Young AY, Anderson J, Tinnel NL, Kornacki M, et al. Expression of the common acute lymphoblastic leukemia antigen (CALLA) on the surface of individual cells of human lymphoblastoid lines. Journal of immunology. 1986 Jan;136(1):320–5. [PubMed] [Google Scholar]
  • 67.Semenza GL. Molecular mechanisms mediating metastasis of hypoxic breast cancer cells. Trends in molecular medicine. 2012 Sep;18(9):534–43. doi: 10.1016/j.molmed.2012.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Scortegagna M, Ding K, Zhang Q, Oktay Y, Bennett MJ, Bennett M, et al. HIF-2alpha regulates murine hematopoietic development in an erythropoietin-dependent manner. Blood. 2005 Apr 15;105(8):3133–40. doi: 10.1182/blood-2004-05-1695. [DOI] [PubMed] [Google Scholar]
  • 69.Percy MJ, Furlow PW, Lucas GS, Li X, Lappin TR, McMullin MF, et al. A gain-of-function mutation in the HIF2A gene in familial erythrocytosis. The New England journal of medicine. 2008 Jan 10;358(2):162–8. doi: 10.1056/NEJMoa073123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Percy MJ, Beer PA, Campbell G, Dekker AW, Green AR, Oscier D, et al. Novel exon 12 mutations in the HIF2A gene associated with erythrocytosis. Blood. 2008 Jun 1;111(11):5400–2. doi: 10.1182/blood-2008-02-137703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kapitsinou PP, Liu Q, Unger TL, Rha J, Davidoff O, Keith B, et al. Hepatic HIF-2 regulates erythropoietic responses to hypoxia in renal anemia. Blood. 2010 Oct 21;116(16):3039–48. doi: 10.1182/blood-2010-02-270322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lee FS, Percy MJ. The HIF pathway and erythrocytosis. Annual review of pathology. 2011;6:165–92. doi: 10.1146/annurev-pathol-011110-130321. [DOI] [PubMed] [Google Scholar]
  • 73.Tojo Y, Sekine H, Hirano I, Pan X, Souma T, Tsujita T, et al. Hypoxia Signaling Cascade for Erythropoietin Production in Hepatocytes. Molecular and cellular biology. 2015 Aug 1;35(15):2658–72. doi: 10.1128/MCB.00161-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rankin EB, Wu C, Khatri R, Wilson TL, Andersen R, Araldi E, et al. The HIF signaling pathway in osteoblasts directly modulates erythropoiesis through the production of EPO. Cell. 2012 Mar 30;149(1):63–74. doi: 10.1016/j.cell.2012.01.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Souma T, Nezu M, Nakano D, Yamazaki S, Hirano I, Sekine H, et al. Erythropoietin Synthesis in Renal Myofibroblasts Is Restored by Activation of Hypoxia Signaling. Journal of the American Society of Nephrology : JASN. 2015 Jun;:8. doi: 10.1681/ASN.2014121184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Laitala A, Aro E, Walkinshaw G, Maki JM, Rossi M, Heikkila M, et al. Transmembrane prolyl 4-hydroxylase is a fourth prolyl 4-hydroxylase regulating EPO production and erythropoiesis. Blood. 2012 Oct 18;120(16):3336–44. doi: 10.1182/blood-2012-07-441824. [DOI] [PubMed] [Google Scholar]
  • 77.Flamme I, Oehme F, Ellinghaus P, Jeske M, Keldenich J, Thuss U. Mimicking hypoxia to treat anemia: HIF-stabilizer BAY 85-3934 (Molidustat) stimulates erythropoietin production without hypertensive effects. PloS one. 2014;9(11):e111838. doi: 10.1371/journal.pone.0111838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Koditz J, Nesper J, Wottawa M, Stiehl DP, Camenisch G, Franke C, et al. Oxygen-dependent ATF-4 stability is mediated by the PHD3 oxygen sensor. Blood. 2007 Nov 15;110(10):3610–7. doi: 10.1182/blood-2007-06-094441. [DOI] [PubMed] [Google Scholar]
  • 79.Cummins EP, Berra E, Comerford KM, Ginouves A, Fitzgerald KT, Seeballuck F, et al. Prolyl hydroxylase-1 negatively regulates IkappaB kinase-beta, giving insight into hypoxia-induced NFkappaB activity. Proceedings of the National Academy of Sciences of the United States of America. 2006 Nov 28;103(48):18154–9. doi: 10.1073/pnas.0602235103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hiwatashi Y, Kanno K, Takasaki C, Goryo K, Sato T, Torii S, et al. PHD1 interacts with ATF4 and negatively regulates its transcriptional activity without prolyl hydroxylation. Experimental cell research. 2011 Dec 10;317(20):2789–99. doi: 10.1016/j.yexcr.2011.09.005. [DOI] [PubMed] [Google Scholar]
  • 81.Gilkes DM, Semenza GL. Role of hypoxia-inducible factors in breast cancer metastasis. Future oncology. 2013 Nov;9(11):1623–36. doi: 10.2217/fon.13.92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xu M, Nagati JS, Xie J, Li J, Walters H, Moon YA, et al. An acetate switch regulates stress erythropoiesis. Nature medicine. 2014 Sep;20(9):1018–26. doi: 10.1038/nm.3587. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Chen R, Xu M, Nagati JS, Hogg RT, Das A, Gerard RD, et al. The Acetate/ACSS2 Switch Regulates HIF-2 Stress Signaling in the Tumor Cell Microenvironment. PloS one. 2015;10(2):e0116515. doi: 10.1371/journal.pone.0116515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Livnah O, Stura EA, Middleton SA, Johnson DL, Jolliffe LK, Wilson IA. Crystallographic evidence for preformed dimers of erythropoietin receptor before ligand activation. Science. 1999 Feb 12;283(5404):987–90. doi: 10.1126/science.283.5404.987. [DOI] [PubMed] [Google Scholar]
  • 85.Pelletier S, Gingras S, Funakoshi-Tago M, Howell S, Ihle JN. Two domains of the erythropoietin receptor are sufficient for Jak2 binding/activation and function. Molecular and cellular biology. 2006 Nov;26(22):8527–38. doi: 10.1128/MCB.01035-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Zhang YL, Radhakrishnan ML, Lu X, Gross AW, Tidor B, Lodish HF. Symmetric signaling by an asymmetric 1 erythropoietin: 2 erythropoietin receptor complex. Molecular cell. 2009 Jan 30;33(2):266–74. doi: 10.1016/j.molcel.2008.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Song KE, Byeon J, Moon DB, Kim HH, Choi YJ, Suh JK. Structural identification of modified amino acids on the interface between EPO and its receptor from EPO BRP, human recombinant erythropoietin by LC/MS analysis. Molecules and cells. 2014 Nov;37(11):819–26. doi: 10.14348/molcells.2014.0214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Elliott S, Lorenzini T, Chang D, Barzilay J, Delorme E. Mapping of the active site of recombinant human erythropoietin. Blood. 1997 Jan 15;89(2):493–502. [PubMed] [Google Scholar]
  • 89.Liu Z, Stoll VS, Devries PJ, Jakob CG, Xie N, Simmer RL, et al. A potent erythropoietin-mimicking human antibody interacts through a novel binding site. Blood. 2007 Oct 1;110(7):2408–13. doi: 10.1182/blood-2007-04-083998. [DOI] [PubMed] [Google Scholar]
  • 90.Lacy SE, DeVries PJ, Xie N, Fung E, Lesniewski RR, Reilly EB. The potency of erythropoietin-mimic antibodies correlates inversely with affinity. Journal of immunology. 2008 Jul 15;181(2):1282–7. doi: 10.4049/jimmunol.181.2.1282. [DOI] [PubMed] [Google Scholar]
  • 91.Casadevall N, Nataf J, Viron B, Kolta A, Kiladjian JJ, Martin-Dupont P, et al. Pure red-cell aplasia and antierythropoietin antibodies in patients treated with recombinant erythropoietin. The New England journal of medicine. 2002 Feb 14;346(7):469–75. doi: 10.1056/NEJMoa011931. [DOI] [PubMed] [Google Scholar]
  • 92.Aoki K, Ono Y, Tabata S, Matsushita A, Ishikawa T. Successful treatment of anti-erythropoietin antibody-mediated pure red cell aplasia with low-dose prednisolone. International journal of hematology. 2013 Feb;97(2):272–4. doi: 10.1007/s12185-013-1258-3. [DOI] [PubMed] [Google Scholar]
  • 93.Matsuki E, Miyakawa Y, Yamane A, Okamoto S. Humanized VB22B minibody for human Mpl stimulates human megakaryopoiesis but does not enhance platelet aggregation. Experimental hematology. 2011 Aug;39(8):829–36. doi: 10.1016/j.exphem.2011.05.001. [DOI] [PubMed] [Google Scholar]
  • 94.Wrighton NC, Farrell FX, Chang R, Kashyap AK, Barbone FP, Mulcahy LS, et al. Small peptides as potent mimetics of the protein hormone erythropoietin. Science. 1996 Jul 26;273(5274):458–64. doi: 10.1126/science.273.5274.458. [DOI] [PubMed] [Google Scholar]
  • 95.Barbone FP, Johnson DL, Farrell FX, Collins A, Middleton SA, McMahon FJ, et al. New epoetin molecules and novel therapeutic approaches. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association. 1999;14(Suppl 2):80–4. doi: 10.1093/ndt/14.suppl_2.80. [DOI] [PubMed] [Google Scholar]
  • 96.Bugelski PJ, Capocasale RJ, Makropoulos D, Marshall D, Fisher PW, Lu J, et al. CNTO 530: molecular pharmacology in human UT-7EPO cells and pharmacokinetics and pharmacodynamics in mice. Journal of biotechnology. 2008 Mar 20;134(1-2):171–80. doi: 10.1016/j.jbiotec.2007.12.005. [DOI] [PubMed] [Google Scholar]
  • 97.Perez-Ruixo JJ, Krzyzanski W, Bouman-Thio E, Miller B, Jang H, Bai SA, et al. Pharmacokinetics and pharmacodynamics of the erythropoietin Mimetibody construct CNTO 528 in healthy subjects. Clinical pharmacokinetics. 2009;48(9):601–13. doi: 10.2165/11317190-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 98.Sathyanarayana P, Houde E, Marshall D, Volk A, Makropoulos D, Emerson C, et al. CNTO 530 functions as a potent EPO mimetic via unique sustained effects on bone marrow proerythroblast pools. Blood. 2009 May 14;113(20):4955–62. doi: 10.1182/blood-2008-08-172320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Makropoulos DA, Achuthanandam R, Avery J, Wilson K, Brosnan K, Miller A, et al. CNTO 530 increases expression of HbA and HbF in murine models of beta-thalassemia and sickle cell anemia. Current pharmaceutical biotechnology. 2013;14(2):242–8. doi: 10.2174/138920113805219449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Fishbane S, Schiller B, Locatelli F, Covic AC, Provenzano R, Wiecek A, et al. Peginesatide in patients with anemia undergoing hemodialysis. The New England journal of medicine. 2013 Jan 24;368(4):307–19. doi: 10.1056/NEJMoa1203165. [DOI] [PubMed] [Google Scholar]
  • 101.Macdougall IC, Rossert J, Casadevall N, Stead RB, Duliege AM, Froissart M, et al. A peptide-based erythropoietin-receptor agonist for pure red-cell aplasia. The New England journal of medicine. 2009 Nov 5;361(19):1848–55. doi: 10.1056/NEJMoa074037. [DOI] [PubMed] [Google Scholar]
  • 102.Bennett CL, Jacob S, Hymes J, Usvyat LA, Maddux FW. Anaphylaxis and hypotension after administration of peginesatide. The New England journal of medicine. 2014 May 22;370(21):2055–6. doi: 10.1056/NEJMc1400883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Zhang J, Randall MS, Loyd MR, Li W, Schweers RL, Persons DA, et al. Role of erythropoietin receptor signaling in Friend virus-induced erythroblastosis and polycythemia. Blood. 2006 Jan 1;107(1):73–8. doi: 10.1182/blood-2005-05-1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lu X, Gross AW, Lodish HF. Active conformation of the erythropoietin receptor: random and cysteine-scanning mutagenesis of the extracellular juxtamembrane and transmembrane domains. The Journal of biological chemistry. 2006 Mar 17;281(11):7002–11. doi: 10.1074/jbc.M512638200. [DOI] [PubMed] [Google Scholar]
  • 105.Cammett TJ, Jun SJ, Cohen EB, Barrera FN, Engelman DM, Dimaio D. Construction and genetic selection of small transmembrane proteins that activate the human erythropoietin receptor. Proceedings of the National Academy of Sciences of the United States of America. 2010 Feb 23;107(8):3447–52. doi: 10.1073/pnas.0915057107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Erickson-Miller CL, DeLorme E, Tian SS, Hopson CB, Stark K, Giampa L, et al. Discovery and characterization of a selective, nonpeptidyl thrombopoietin receptor agonist. Experimental hematology. 2005 Jan;33(1):85–93. doi: 10.1016/j.exphem.2004.09.006. [DOI] [PubMed] [Google Scholar]
  • 107.Nurden AT, Viallard JF, Nurden P. New-generation drugs that stimulate platelet production in chronic immune thrombocytopenic purpura. Lancet. 2009 May 2;373(9674):1562–9. doi: 10.1016/S0140-6736(09)60255-5. [DOI] [PubMed] [Google Scholar]
  • 108.Kim YK, Lee SS, Jeong SH, Ahn JS, Yang DH, Lee JJ, et al. Efficacy and safety of eltrombopag in adult refractory immune thrombocytopenia. Blood research. 2015 Mar;50(1):19–25. doi: 10.5045/br.2015.50.1.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Erickson-Miller CL, Delorme E, Tian SS, Hopson CB, Landis AJ, Valoret EI, et al. Preclinical activity of eltrombopag (SB-497115), an oral, nonpeptide thrombopoietin receptor agonist. Stem cells. 2009 Feb;27(2):424–30. doi: 10.1634/stemcells.2008-0366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kautz L, Jung G, Valore EV, Rivella S, Nemeth E, Ganz T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nature genetics. 2014 Jul;46(7):678–84. doi: 10.1038/ng.2996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Kautz L, Jung G, Nemeth E, Ganz T. Erythroferrone contributes to recovery from anemia of inflammation. Blood. 2014 Oct 16;124(16):2569–74. doi: 10.1182/blood-2014-06-584607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Rochette L, Gudjoncik A, Guenancia C, Zeller M, Cottin Y, Vergely C. The iron-regulatory hormone hepcidin: a possible therapeutic target? Pharmacology & therapeutics. 2015 Feb;146:35–52. doi: 10.1016/j.pharmthera.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 113.Fibach E, Rachmilewitz E. The role of oxidative stress in hemolytic anemia. Current molecular medicine. 2008 Nov;8(7):609–19. doi: 10.2174/156652408786241384. [DOI] [PubMed] [Google Scholar]
  • 114.Scheibye-Knudsen M, Fang EF, Croteau DL, Wilson DM, 3rd, Bohr VA. Protecting the mitochondrial powerhouse. Trends in cell biology. 2015 Mar;25(3):158–70. doi: 10.1016/j.tcb.2014.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Prus E, Fibach E. Effect of iron chelators on labile iron and oxidative status of thalassaemic erythroid cells. Acta haematologica. 2010;123(1):14–20. doi: 10.1159/000258958. [DOI] [PubMed] [Google Scholar]
  • 116.Dev A, Byrne SM, Verma R, Ashton-Rickardt PG, Wojchowski DM. Erythropoietin-directed erythropoiesis depends on serpin inhibition of erythroblast lysosomal cathepsins. The Journal of experimental medicine. 2013 Feb 11;210(2):225–32. doi: 10.1084/jem.20121762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sbongile M, Soliman ME. In silico identification of irreversible cathepsin B inhibitors as anti-cancer agents: virtual screening, covalent docking analysis and molecular dynamics simulations. Combinatorial chemistry & high throughput screening. 2015;18(4):399–410. doi: 10.2174/1386207318666150305154621. [DOI] [PubMed] [Google Scholar]
  • 118.Nabinger SC, Chan RJ. Shp2 function in hematopoietic stem cell biology and leukemogenesis. Current opinion in hematology. 2012 Jul;19(4):273–9. doi: 10.1097/MOH.0b013e328353c6bf. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chong ZZ, Maiese K. The Src homology 2 domain tyrosine phosphatases SHP-1 and SHP-2: diversified control of cell growth, inflammation, and injury. Histology and histopathology. 2007 Nov;22(11):1251–67. doi: 10.14670/hh-22.1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wojchowski DM, Su S, Johnson A, Jachimowicz E, Stokes MP, Green JM, et al. Discovery of novel EPO/EPOR/JAK2 targets and signal transduction factors via proteomic-based interrogations of post-translational motif modifications. American Society of Hematology Annual Meeting Proceedings: Blood. 2014:1335. [Google Scholar]
  • 121.Li S, Hsu DD, Wang H, Feng GS. Dual faces of SH2-containing protein-tyrosine phosphatase Shp2/PTPN11 in tumorigenesis. Frontiers of medicine. 2012 Sep;6(3):275–9. doi: 10.1007/s11684-012-0216-4. [DOI] [PubMed] [Google Scholar]
  • 122.Usenko T, Chan G, Torlakovic E, Klingmuller U, Neel BG. Leukemogenic Ptpn11 allele causes defective erythropoiesis in mice. PloS one. 2014;9(10):e109682. doi: 10.1371/journal.pone.0109682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Sharma N, Kumar V, Everingham S, Mali RS, Kapur R, Zeng LF, et al. SH2 domain-containing phosphatase 2 is a critical regulator of connective tissue mast cell survival and homeostasis in mice. Molecular and cellular biology. 2012 Jul;32(14):2653–63. doi: 10.1128/MCB.00308-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sharma N, Everingham S, Zeng LF, Zhang ZY, Kapur R, Craig AW. Oncogenic KIT-induced aggressive systemic mastocytosis requires SHP2/PTPN11 phosphatase for disease progression in mice. Oncotarget. 2014 Aug 15;5(15):6130–41. doi: 10.18632/oncotarget.2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Poole AW, Jones ML. A SHPing tale: perspectives on the regulation of SHP-1 and SHP-2 tyrosine phosphatases by the C-terminal tail. Cellular signalling. 2005 Nov;17(11):1323–32. doi: 10.1016/j.cellsig.2005.05.016. [DOI] [PubMed] [Google Scholar]
  • 126.Van Zant G, Shultz L. Hematologic abnormalities of the immunodeficient mouse mutant, viable motheaten (mev) Experimental hematology. 1989 Feb;17(2):81–7. [PubMed] [Google Scholar]
  • 127.Klingmuller U, Lorenz U, Cantley LC, Neel BG, Lodish HF. Specific recruitment of SH-PTP1 to the erythropoietin receptor causes inactivation of JAK2 and termination of proliferative signals. Cell. 1995 Mar 10;80(5):729–38. doi: 10.1016/0092-8674(95)90351-8. [DOI] [PubMed] [Google Scholar]
  • 128.Wang HM, Xu YF, Ning SL, Yang DX, Li Y, Du YJ, et al. The catalytic region and PEST domain of PTPN18 distinctly regulate the HER2 phosphorylation and ubiquitination barcodes. Cell research. 2014 Sep;24(9):1067–90. doi: 10.1038/cr.2014.99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Huang WQ, Lin Q, Zhuang X, Cai LL, Ruan RS, Lu ZX, et al. Structure, function, and pathogenesis of SHP2 in developmental disorders and tumorigenesis. Current cancer drug targets. 2014;14(6):567–88. doi: 10.2174/1568009614666140717105001. [DOI] [PubMed] [Google Scholar]
  • 130.He RJ, Yu ZH, Zhang RY, Zhang ZY. Protein tyrosine phosphatases as potential therapeutic targets. Acta pharmacologica Sinica. 2014 Oct;35(10):1227–46. doi: 10.1038/aps.2014.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chio CM, Lim CS, Bishop AC. Targeting a cryptic allosteric site for selective inhibition of the oncogenic protein tyrosine phosphatase Shp2. Biochemistry. 2015 Jan 20;54(2):497–504. doi: 10.1021/bi5013595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Geyer HL, Mesa RA. Therapy for myeloproliferative neoplasms: when, which agent, and how? Blood. 2014 Dec 4;124(24):3529–37. doi: 10.1182/blood-2014-05-577635. [DOI] [PubMed] [Google Scholar]
  • 133.Wang LJ, Jiang B, Wu N, Wang SY, Shi DY. Small molecules as potent protein tyrosine phosphatase 1B (PTP1B) inhibitors documented in patents from 2009 to 2013. Mini reviews in medicinal chemistry. 2015;15(2):104–22. doi: 10.2174/1389557515666150203144339. [DOI] [PubMed] [Google Scholar]
  • 134.Hou X, Li R, Li K, Yu X, Sun JP, Fang H. Fast identification of novel lymphoid tyrosine phosphatase inhibitors using target-ligand interaction-based virtual screening. Journal of medicinal chemistry. 2014 Nov 26;57(22):9309–22. doi: 10.1021/jm500692u. [DOI] [PubMed] [Google Scholar]
  • 135.Low JL, Chai CL, Yao SQ. Bidentate inhibitors of protein tyrosine phosphatases. Antioxidants & redox signaling. 2014 May 10;20(14):2225–50. doi: 10.1089/ars.2013.5710. [DOI] [PubMed] [Google Scholar]
  • 136.Peslak SA, Wenger J, Bemis JC, Kingsley PD, Koniski AD, McGrath KE, et al. EPO-mediated expansion of late-stage erythroid progenitors in the bone marrow initiates recovery from sublethal radiation stress. Blood. 2012 Sep 20;120(12):2501–11. doi: 10.1182/blood-2011-11-394304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Dussiot M, Maciel TT, Fricot A, Chartier C, Negre O, Veiga J, et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in beta-thalassemia. Nature medicine. 2014 Apr;20(4):398–407. doi: 10.1038/nm.3468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Guihard S, Clay D, Cocault L, Saulnier N, Opolon P, Souyri M, et al. The MAPK ERK1 is a negative regulator of the adult steady-state splenic erythropoiesis. Blood. 2010 May 6;115(18):3686–94. doi: 10.1182/blood-2009-09-242487. [DOI] [PubMed] [Google Scholar]
  • 139.Wu DC, Paulson RF. Hypoxia regulates BMP4 expression in the murine spleen during the recovery from acute anemia. PloS one. 2010;5(6):e11303. doi: 10.1371/journal.pone.0011303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Harandi OF, Hedge S, Wu DC, McKeone D, Paulson RF. Murine erythroid short-term radioprotection requires a BMP4-dependent, self-renewing population of stress erythroid progenitors. The Journal of clinical investigation. 2010 Dec;120(12):4507–19. doi: 10.1172/JCI41291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Lau CI, Outram SV, Saldana JI, Furmanski AL, Dessens JT, Crompton T. Regulation of murine normal and stress-induced erythropoiesis by Desert Hedgehog. Blood. 2012 May 17;119(20):4741–51. doi: 10.1182/blood-2011-10-387266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.An X, Schulz VP, Li J, Wu K, Liu J, Xue F, et al. Global transcriptome analyses of human and murine terminal erythroid differentiation. Blood. 2014 May 29;123(22):3466–77. doi: 10.1182/blood-2014-01-548305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Paralkar VR, Mishra T, Luan J, Yao Y, Kossenkov AV, Anderson SM, et al. Lineage and species-specific long noncoding RNAs during erythro-megakaryocytic development. Blood. 2014 Mar 20;123(12):1927–37. doi: 10.1182/blood-2013-12-544494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Khoriaty R, Vasievich MP, Jones M, Everett L, Chase J, Tao J, et al. Absence of a red blood cell phenotype in mice with hematopoietic deficiency of SEC23B. Molecular and cellular biology. 2014 Oct 1;34(19):3721–34. doi: 10.1128/MCB.00287-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Verma R, Su S, McCrann DJ, Green JM, Leu K, Young PR, et al. RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development. The Journal of experimental medicine. 2014 Aug 25;211(9):1715–22. doi: 10.1084/jem.20130624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Trakarnsanga K, Wilson MC, Lau W, Singleton BK, Parsons SF, Sakuntanaga P, et al. Induction of adult levels of beta-globin in human erythroid cells that intrinsically express embryonic or fetal globin by transduction with KLF1 and BCL11A-XL. Haematologica. 2014 Nov;99(11):1677–85. doi: 10.3324/haematol.2014.110155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Huang X, Shah S, Wang J, Ye Z, Dowey SN, Tsang KM, et al. Extensive ex vivo expansion of functional human erythroid precursors established from umbilical cord blood cells by defined factors. Molecular therapy : the journal of the American Society of Gene Therapy. 2014 Feb;22(2):451–63. doi: 10.1038/mt.2013.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Saliba J, Hamidi S, Lenglet G, Langlois T, Yin J, Cabagnols X, et al. Heterozygous and homozygous JAK2(V617F) states modeled by induced pluripotent stem cells from myeloproliferative neoplasm patients. PloS one. 2013;8(9):e74257. doi: 10.1371/journal.pone.0074257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. The New England journal of medicine. 2013 Dec 19;369(25):2379–90. doi: 10.1056/NEJMoa1311347. [DOI] [PubMed] [Google Scholar]
  • 150.Asada N, Takase M, Nakamura J, Oguchi A, Asada M, Suzuki N, et al. Dysfunction of fibroblasts of extrarenal origin underlies renal fibrosis and renal anemia in mice. The Journal of clinical investigation. 2011 Oct;121(10):3981–90. doi: 10.1172/JCI57301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Xie L, Pi X, Townley-Tilson WH, Li N, Wehrens XH, Entman ML, et al. PHD2/3-dependent hydroxylation tunes cardiac response to beta-adrenergic stress via phospholamban. The Journal of clinical investigation. 2015 Jul 1;125(7):2759–71. doi: 10.1172/JCI80369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Miyake S, Muramatsu R, Hamaguchi M, Yamashita T. Prolyl hydroxylase regulates axonal rewiring and motor recovery after traumatic brain injury. Cell death & disease. 2015;6:e1638. doi: 10.1038/cddis.2015.5. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES