Next Article in Journal
The Role of Nondiabetic Hyperglycemia in Critically Ill Patients with Acute Ischemic Stroke
Next Article in Special Issue
Special Issue: “Emerging Therapies and Strategies in Thalassemia: Toward a New Era in Management”
Previous Article in Journal
Variation in Tap Water Mineral Content in the United Kingdom: Is It Relevant for Kidney Stone Disease?
Previous Article in Special Issue
Safety and Efficacy of the New Combination Iron Chelation Regimens in Patients with Transfusion-Dependent Thalassemia and Severe Iron Overload
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Does Hepcidin Tuning Have a Role among Emerging Treatments for Thalassemia?

1
Thalassemia Reference Centre, 10043 Orbassano, Italy
2
Regional HUB Centre for Thalassaemia and Haemoglobinopathies, Department of Medicine, Azienda Ospedaliero Universitaria S. Anna, 44124 Ferrara, Italy
3
University of Torino, 10043 Torino, Italy
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2022, 11(17), 5119; https://doi.org/10.3390/jcm11175119
Submission received: 17 June 2022 / Revised: 21 August 2022 / Accepted: 27 August 2022 / Published: 30 August 2022

Abstract

:
The treatments available for thalassemia are rapidly evolving, with major advances made in gene therapy and the modulation of erythropoiesis. The latter includes the therapeutic potential of hepcidin tuning. In thalassemia, hepcidin is significantly depressed, and any rise in hepcidin function has a positive effect on both iron metabolism and erythropoiesis. Synthetic hepcidin and hepcidin mimetics have been developed to the stage of clinical trials. However, they have failed to produce an acceptable efficacy/safety profile. It seems difficult to avoid iron over-restricted erythropoiesis when directly using hepcidin as a drug. Indirect approaches, each one with their advantages and disadvantages, are many and in full development. The ideal approach is to target erythroferrone, the main inhibitor of hepcidin expression, the plasma concentrations of which are greatly increased in iron-loading anemias. Potential means of improving hepcidin function in thalassemia also include acting on TMPRSS6, TfR1, TfR2 or ferroportin, the target of hepcidin. Only having a better understanding of the crosslinks between iron metabolism and erythropoiesis will elucidate the best single option. In the meantime, many potential combinations are currently being explored in preclinical studies. Any long-term clinical study on this approach should include the wide monitoring of functions, as the effects of hepcidin and its modulators are not limited to iron metabolism and erythropoiesis. It is likely that some of the aspects of hepcidin tuning described briefly in this review will play a role in the future treatment of thalassemia.

1. Introduction

Thalassemia and Sickle Cell Disease (SCD) are among the most common inherited disorders, affecting millions of people worldwide [1,2].
β-thalassemia is caused by mutations in the β-globin gene that lead to unbalanced α- and β-chain production, with the expansion of abnormal erythroid precursors, maturation blockage and increased apoptosis of late erythroblasts (Figure 1). This detrimental condition of ineffective erythropoiesis (IE) is tightly linked to iron metabolism dysregulation with hepcidin suppression, which predisposes patients to severe iron loading even in the absence of transfusions [3]. Thalassemia is the prototype of the so-called iron-loading anemias.
The degree of anemia and IE influence different phenotypes. Thalassemia major (TM) is defined as a severe form of anemia with early bone alterations and lifelong transfusion need; thalassemia intermedia (TI) includes a broad spectrum of severity, from very mild conditions to moderate ones with a high risk of clinical complications. Regarding basic treatment, thalassemias are grouped as transfusion-dependent β-thalassemia (TDT) and non-transfusion-dependent β-thalassemia (NTDT) [4]. The TM/TI and TDT/NTDT categories are not synonymous but complementary; experienced clinicians label a patient permanently as TM or TI, according to the clinical presentation and the severity of genetic defects. On the contrary, the TDT/NTDT label may vary according to local blood availability and individual clinical course. A significant proportion of TI patients move sooner or later from NTDT to TDT due to worsening anemia or for preventing/controlling complications. In general, both TDT and most NTDT patients suffer from iron overload, which requires chronic iron chelation therapy to prevent iron-related complications, such as endocrinopathies and liver and heart disease [4].
Sickle Cell Disease (SCD) is a monogenic disorder that results in progressive multiorgan disease due to intravascular hemolysis, acute vaso-occlusive crisis and chronic inflammation. If, in SCD, on the one hand, anemia leads to the suppression of hepcidin, on the other hand, inflammation and chronic hemolysis upregulate hepcidin. The final result of these two patterns in each patient may vary widely, but a minority of SCD patients have a pattern of iron-loading anemias [5].

2. Hepcidin–Ferroportin Axis

Hepcidin is a peptide hormone produced by the HAMP gene (19q13), initially associated with in vitro antimicrobial activity only [6,7]. Then, hepcidin emerged as the key hormone for iron homeostasis regulation [8]. Encoded as a pro-peptide by the cleavage of two isoforms, hepcidin-25 represents the active one, with an N-terminal residue of five amino acids that are crucial for iron regulation, inhibition of dietary iron absorption and iron release from storage [9]. Hepcidin is primarily synthesized in the hepatocytes and, to a lesser extent, by monocytes [10], macrophages [11], adipocytes [12], the kidneys [13], the small intestine, the placenta [14], retinal cells [15] and cardiomyocytes [16]. Hepcidin is upregulated by iron levels [17], inflammation and infection [18,19], while anemia, hypoxia, iron deficiency, ineffective erythropoiesis and increased erythropoietin (EPO) levels are down-regulating factors [19,20]. Hepcidin synthesis is mainly controlled throughout the bone morphogenetic proteins BMP6 and, to a lesser extent, BMP2 produced by endothelial cells, which activate signal transmission through SMAD proteins (Figure 1) [21,22,23]. Once secreted into the circulation, hepcidin binds to ferroportin (FPN), the only known cellular iron exporter in vertebrates, localized at the basolateral membranes of duodenal enterocytes [24]. FPN is also abundantly expressed on cells that recycle iron from senescent red blood cells, such as macrophages of the spleen, bone marrow and liver [24]. Hepcidin binding to ferroportin is coupled to iron binding, with an 80-fold increase in hepcidin affinity in the presence of iron [25]. Hepcidin binding occludes the central cavity of FPN, interfering with iron export independently of endocytosis [26]. The iron-related differential affinity for FPN has the important function of determining the inhibition of cells actively involved in iron export, such as intestinal cells and macrophages [25]. After hepcidin binding, FPN is internalized and degraded by lysosomes, requiring the resynthesis of degraded ferroportin molecules. The final result is that the outflow of iron from cells is reduced, and the intracellular storage increases [27]. Hepcidin’s effect lasts up to 48 h.
The massive erythron expansion of thalassemia requires up to 100-fold the physiological daily iron amount [28,29]. This requirement is met by the increased expression of erythroferrone (Erfe), a recently identified iron-regulatory hormone highly expressed in erythroblasts in response to EPO stimulation [30]. Erfe-knockout mice are not anemic, indicating that Erfe has a modest effect on hepcidin repression in steady state. However, Erfe contributes to iron loading in mice with β-thalassemia [31]. Erfe acts as a potent hepcidin suppression, thus enhancing duodenal iron absorption and iron mobilization from stores. This favors iron availability for the increased erythropoietic demand. The erythroid sensing, throughout Erfe, prevails over the iron sensing. This aspect is useful in physiological conditions, but, in thalassemias, it is responsible for triggering iron overload, both in NTDT and TDT patients [20,31,32,33].
The most potent Erfe inhibitor is the liver transmembrane serine protease TMPRSS6 (transmembrane serine protease 6) or matriptase-2 TMPRSS6, which is mutated in patients with iron-refractory iron deficiency anemia (IRIDA) [34]. TMPRSS6 cleaves the BMP coreceptor hemojuvelin (HJV), thereby avoiding the overactivation of BMP/SMAD signaling and hepcidin transcription [35].
The expansion of the erythropoietic compartment is backed by the expression of transferrin receptor 1 (TfR1) and transferrin receptor 2 (TfR2), which modulate the iron supply to the erythron [36]. TfR1 is the most abundant cell membrane protein on erythroblasts and is essential in mediating transferrin endocytosis.
Unfortunately, a simple and direct marker of erythropoiesis is lacking. This makes intra- and interpatient comparisons, with or without treatment, challenging in thalassemia. Recently, the ratio of sTfR1 plasma concentration (proportional to the volume of erythroid tissue) divided by absolute reticulocyte count (proportional to effective erythroid output from the bone marrow) has been proposed as the best index to quantify ineffective erythropoiesis [37]. In mouse models of beta-thalassemia, decreased expression of TfR1 has been associated with improved iron metabolism and IE [38]. TfR2, a constitutive component of the Epo receptor complex, is expressed during the differentiation of erythroid cells [39], but its function has not been fully unveiled. Even so, TfR2 inhibition has potential therapeutic implications as, in thalassemic mice, the knockout of TfR2 lowers Epo and Erfe levels, increasing the proportion of mature precursors and hemoglobin [40].
Thalassemia is the prototype of iron-loading anemias. Urinary hepcidin levels are suppressed in patients with NTDT not receiving transfusions. Hepcidin is also low, but to a less pronounced extent, in patients on a regular transfusion program, even with more severe iron overload [41]. This finding is not surprising, as transfusion improves anemia and suppresses erythropoiesis, with an increase in hepcidin production [42]. However, hepcidin concentrations progressively decrease in the intervals between transfusions [43,44]. In summary, any severe form of thalassemia, even on regular transfusion, should be considered an iron-loading anemia.

3. Targeting the Hepcidin–Ferroportin Axis in Thalassemia

Restoration of hepcidin levels in thalassemia could reduce iron absorption, improve ineffective erythropoiesis and possibly, in the long term, also reduce iron toxicity [45]. Proof-of-principle studies indicate that a moderate increase in hepcidin expression reduces tissue iron levels, with a beneficial effect on erythropoiesis, in an NTDT mouse model [46].

3.1. Hepcidin Mimetics

Two hepcidin mimetics (LJPC-401 and PTG-300, see Table 1) have been developed independently and have demonstrated efficacy in preclinical models. Phase 1 studies on humans showed a significant and long-lasting reduction in serum iron following their administration [47]. LJPC-401 is a synthetic, full-length hepcidin mimetic with the mature form of the human hepcidin. It reached a phase 2 study, primarily aiming at evaluating its efficacy in cardiac iron reduction in adult TDT patients with myocardial iron overload (NCT03381833). The study failed to produce an acceptable efficacy/safety profile and further development was foregone. A slightly different approach was applied to synthesize PTG-300, a hepcidin mimetic that shares only the N-terminal portion of the human peptide sequence [48]. PTG-300 was investigated in a phase 2 clinical trial on TDT and NTDT patients (NCT03802201) that confirmed its ability in reducing transferrin saturation and serum iron [49]. Unfortunately, for PTG-300, injection site reactions have been a significant limitation. The so-called mini-hepcidins are truncated hepcidin peptides based on the N-terminal sequence of hepcidin and have several potential advantages, but, at present, none has entered clinical development. However, preclinical results in an NTDT mouse model are promising [50]. In addition, in a new mouse model with a severe transfusion-dependent thalassemia disease phenotype, a mini-hepcidin (MH), combined with red blood cell transfusion, ameliorated IE, splenomegaly and cardiac iron overload [51]. Recently, new mini-hepcidins with improved drug-like properties have been designed using head-to-tail cyclization and N-methylation [52].

3.2. Hepcidin Agonists

Another approach to the search for hepcidin agonist action is indirect, by inhibiting the main regulators of hepcidin function, such as TMPRSS6 and Erfe, or by inhibiting the hepcidin target ferroportin [23].
  • TMPRSS6 inhibition
In an NTDT mouse model, loss of TMPRSS6 significantly attenuated the disease phenotype [53]. Two independent studies, using oligonucleotides such as antisense oligonucleotides (ASO) or small interfering RNA (siRNA), have confirmed these findings, not only decreasing iron loading but also improving erythropoiesis, splenomegaly and anemia [54,55]. Two phase 2 clinical trials on NTDT patients are currently ongoing, using IONIS TMPRSS6-LRx (NCT04059406) and SLN124 (NCT04718844). Recently, the anti-TMPRSS6 antibody MWTx-003 was presented as a promising therapy for iron overload disorders where an iron restriction is beneficial (Bruxin, ASH 2021).
  • Ferroportin inhibition
Advances in understanding the structure of hepcidin-bound ferroportin and its iron homeostatic mechanisms [25] opened the door to a new approach: targeting ferroportin directly. This offers great potential in tuning the restriction of cellular iron export. Many compounds have been screened for this function. The first to enter development has been vamifeport (VIT-2763), a small molecule orally administered, that acts as an inhibitor of ferroportin, competing with hepcidin for binding to ferroportin [56]. In healthy volunteers, a temporary decrease in mean serum iron levels, transferrin saturation and a shift in mean serum hepcidin peaks followed the administration of VIT-2763 [57]. A phase 2 study of VIT-2763 in NTDT (NCT04364269) is ongoing.
  • Erfe inhibition
In theory, Erfe inhibition is the best approach to tune hepcidin function for iron-loading anemias, because the function of Erfe is to tune hepcidin merely based on erythroid needs. In a thalassemia mouse model, Erfe inhibition blocked its suppressive effects on hepcidin, with amelioration of the phenotype [31]. Furthermore, due to its distribution and picomolar concentration, Erfe is the ideal target for monoclonal antibodies. This approach, using N-terminal neutralizing antibodies, has been recently applied with promising results in thalassemia mice [58].
  • TfR2 inactivation
The inactivation of the EPO receptor partner, TfR2, in a knockout thalassemia intermedia mouse model improved erythropoiesis and red blood cell morphology, as well as anemia and iron overload [40]. However, the beneficial effects became attenuated over time, possibly due to insufficient iron availability to sustain the enhanced erythropoiesis. Germline deletion of TfR2, including haploinsufficiency, has a similar impact in the thalassemic model [40]. Data from TfR2-haploinsufficient thalassemic mice suggest that TfR2-specific targeting by antisense oligonucleotides or small interfering RNAs has great therapeutic potential in NTDT [59]. A better understanding of the TfR2–EPOR interaction may lead to the design of interfering molecules, mimicking an erythroid-specific TfR2 depletion. Unlike erythropoiesis-stimulating agents, the TfR2 approach enhances EPO-mediated effects exclusively in erythroid cells, with potential advantages for long-term safety [59].
  • Apotransferrin
In thalassemic erythropoiesis, iron is abundant due to the massive erythroid demand and hepcidin suppression, but is underutilized due to the genetic defect of hemoglobinization. As the iron turnover is high, the therapeutic use of apotransferrin has been hypothesized. In NTDT mice, apotransferrin infusion improved anemia, splenomegaly and plasma EPO, decreased membrane-associated α- globin precipitates and normalized the RBC half-life [38]. In addition, a reduction in cardiosiderosis has also been observed [60]. A phase 2 clinical trial using i.v. infusion of apotransferrin every other week is currently being performed in beta-thalassemia (NCT03993613).

3.3. Combination Therapy

The availability of several therapeutic tools targeting different pathways has allowed the exploration of balanced combinations, searching for a potential additive or even synergistic effect (Table 2). For example, in NTDT mice, TMPRSS6 inhibition (by TMPRSS6-ASO or siRNA) combined with the iron-chelating agent deferiprone produced some additive effects on the improvement of erythropoiesis and iron overload [61,62,63]. Another interesting approach has been assessed by combining TMPRSS6 inhibition with EPO administration or removing a single TfR2 allele in the bone marrow of NTDT mice. Both combinations were more effective than a single agent in ameliorating anemia and splenomegaly [59]. Moreover, the inhibition of both TMPRSS6 and TfR2 gave interesting results in Hbbth3/+ thalassemic mice: even with the iron overload due to the TfR2 double mutation, a therapeutic effect on both erythropoiesis and anemia was obtained [64]. Another recent approach combined a direct ferroportin inhibitor (VIT-2763) with an iron chelator (deferasirox) [65]. The promising results in NTDT herald potential clinical trials. Of many other possible combinations, several are worthwhile at least in vitro testing, given the potentially strong rationale, such as Erfe inhibition with iron chelation.

4. The Background of Treatment Options

4.1. Conventional Treatment

Severe thalassemia requires regular transfusions and daily iron chelation treatment, accompanied by high-quality monitoring and follow-up. The latter is often enough in TI to preserve good quality of life and prevent long-term thalassemia-related complications such as bone alterations, hypersplenism, thrombotic events, leg ulcers and acquired elastopathy. Where quality of care and patient adherence is kept optimal, the long-term results in terms of survival and quality of life are good, with a life expectancy that approaches normality. However, many literature reports indicate the significant prevalence of long-term complications due to poor adherence and consequent poor control of iron overload-related toxicity.

4.2. Stem Cell Transplantation

A standard approach to the conditioning regimen and immunosuppression has been set for time in different risk classes [66,67]. The likelihood of success in the low-risk category is more than 90% and, differently from the past, it is not limited to pediatric subjects. Therefore, balanced counselling is important to weigh the advantages and disadvantages of conventional treatment versus stem cell transplantation in each individual, especially in the intermediate risk class and according to local health resources.

4.3. Gene Therapy

The gene therapy approach in hemoglobinopathies was introduced many years ago [68], but only in recent times has it seen an impressive acceleration in terms of clinical trials, up to the first approval by regulatory agencies [69]. Today, the classical gene addition is performed by lentiviral vector insertion into stem cells after a myeloablative conditioning regimen [70,71]. Impressive results regarding the efficacy [71] are counterbalanced by long-term safety issues in the light of recent complications with cases of leukemia and myelodysplasia in an SCD trial [72]. The impact of disease-related rather than procedure-related complications is still unclear. Recently, the hypothesis has emerged that the stress hematopoiesis in transplanted cells may drive clonal expansion and the leukemogenic transformation of preexisting premalignant clones [73]. At present, the authors of this review believe that, with the exclusion of ongoing trials, the gene addition approach for hemoglobinopathies will not experience significant development.
On the contrary, the genome editing approach is developing rapidly, with great expectations in many fields of medicine, including hemoglobinopathies. At present, for both thalassemia and Sickle Cell Disease, rather than repairing the causative mutations, the simpler approach of knocking out the lineage-specific regulatory element of the BCL11A gene is under intense investigation. Suppressing BCL11A potently affects fetal hemoglobin reactivation, mitigating or cancelling the pathological impact on both disorders. Among several techniques, CRISPR-Cas9-based genome engineering is by far the most applied one. Several clinical trials are ongoing in thalassemia (NCT03432364, NCT03655678, NCT04208529) and SCD (NCT03745287, NCT04819841), with impressive preliminary results as regards the efficacy. In terms of safety, even if several issues have been solved, concerns associated with the nature of this technique (double strains cut and predisposition to deletions) are still unresolved. It is likely that the full approval of the genome editing approach for treating hemoglobinopathies will require several years of intensive research.
In general, in the case of potential access to gene therapy, the patient needs balanced counselling that takes into account first the access to and quality of conventional treatment. When full access and high quality are available, prudence is due. For a patient living in a country with inadequate care, the likelihood of stem cell transplantation or a gene therapy approach is balanced by the risk of poor conventional treatment. Obviously, these considerations will remain theoretical if the high cost of gene therapy does not lower consistently.

4.4. Modulation of Erythropoiesis and Erythrocyte Metabolism

Given the present limitations of conventional treatment, stem cell transplantation and gene therapy described above, any intervention able to significantly ameliorate the erythropoiesis in hemoglobinopathies is meaningful. Several potential new treatments are emerging from advances in the pathophysiology of thalassemia targeting ineffective erythropoiesis, iron or erythrocyte metabolism (Figure 2).
  • Mitapivat is an oral, small-molecule allosteric activator of pyruvate kinase in red blood cells [74] that has been very recently approved as a treatment for hemolytic anemia in adults with pyruvate kinase (PK) deficiency. PK is functionally normal but low in thalassemic red blood cells, and mitapivat, in a mouse model of thalassemia, demonstrated an interesting improvement in ineffective erythropoiesis and anemia [75]. A phase II study in beta and alpha thalassemia patients showed encouraging results [76]. A clinical trial is ongoing in TDT (NCT04770779). Etavopivat is an analogue compound in development to treat SCD and thalassemia [77].
  • Luspatercept has been recently approved for TDT patients in Europe, the United States and other countries [78,79]. It acts as a trap of the soluble ligands of activin receptor type 2, eventually resulting in more effective RBC differentiation (Figure 1). The clinical result is a consistent reduction in the transfusion burden. Preliminary results from ongoing trials in NTDT indicate a consistent increase in total hemoglobin (NCT04064060).

5. Discussion

As hepcidin has a paramount role in iron homeostasis, its targeting has great potential, especially in conditions where hepcidin is deeply suppressed, as in iron-loading anemias, including thalassemias. As a small peptide, it appears natural to include hepcidin in the long list of synthetic peptide hormones successfully developed and used today as effective drugs in the clinical setting. At present, however, this does not appear likely for hepcidin or hepcidin mimetics, due to challenges in achieving a stable compound, the need for frequent subcutaneous injections and the frequency of site reactions. More importantly, it is difficult to tune their effect, avoiding iron over-restricted erythropoiesis. When the therapeutic window is narrow, the general rule is to turn to indirect tuning. This has been the case for hepcidin agonists, with many studies and interesting results presented regarding single agents and combinations. The first to be used in clinical trials was TMPRSS6, and many other new entries can be expected. As the effects of hepcidin and its modulators are not limited to iron metabolism and erythropoiesis, it is important to develop long-term studies examining potential positive and negative consequences. For example, the trend towards osteoporosis in thalassemia is well known and important. Using hepcidin or Erfe in animals, there have been some negative findings on bone markers that, if confirmed, pose a challenge for their long-term use. It is difficult today to define the precise position of hepcidin tuning in the rapidly expanding pipeline of treatment options for thalassemias. Excluding a curative effect, we can forecast a relevant role in lowering the burden of the disease and improving the phenotype, as is happening for luspatercept, alone and in combination.

6. Conclusions

Hepcidin tuning reached the phase of clinical development thanks to the progress of our understanding on the crosslinks between iron metabolism and erythropoiesis. Advanced clinical trials on different aspects of hepcidin tuning are currently underway. It is likely that this type of approach will play a role in the future treatment of thalassemia.

Author Contributions

Writing—review and editing, F.L. and A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

F.L. received speaker honoraria from Celgene (Bristol Myers Squibb); A.P. received research grants from Acceleron, Celgene (Bristol Myers Squibb), Novartis, Apopharma and Chiesi and consulting fees from Celgene (Bristol Myers Squibb).

References

  1. Piel, F.B.; Steinberg, M.H.; Rees, D.C. Sickle Cell Disease. N. Engl. J. Med. 2017, 376, 1561–1573. [Google Scholar] [CrossRef] [PubMed]
  2. Iolascon, A.; De Franceschi, L.; Muckenthaler, M.; Taher, A.; Rees, D.; De Montalembert, M.; Rivella, S.; Eleftheriou, A.; Cappellini, M.D. EHA Research Roadmap on Hemoglobinopathies and Thalassemia: An Update. HemaSphere 2019, 3, e208. [Google Scholar] [CrossRef] [PubMed]
  3. Cazzola, M. Ineffective erythropoiesis and its treatment. Blood 2021, 139, 2460–2470. [Google Scholar] [CrossRef]
  4. Taher, A.T.; Musallam, K.M.; Cappellini, M.D. β-Thalassemias. N. Engl. J. Med. 2021, 384, 727–743. [Google Scholar] [CrossRef]
  5. Porter, J.B. Pathophysiology of transfusional iron overload: Contrasting patterns in thalassemia major and sickle cell disease. Hemoglobin 2009, 33 (Suppl. 1), S37–S45. [Google Scholar] [CrossRef] [PubMed]
  6. Krause, A.; Neitz, S.; Mägert, H.J.; Schulz, A.; Forssmann, W.G.; Schulz-Knappe, P.; Adermann, K. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett. 2000, 480, 147–150. [Google Scholar] [CrossRef]
  7. Park, C.H.; Valore, E.V.; Waring, A.J.; Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001, 276, 7806–7810. [Google Scholar] [CrossRef] [PubMed]
  8. Ganz, T. Erythropoietic regulators of iron metabolism. Free Radic. Biol. Med. 2019, 133, 69–74. [Google Scholar] [CrossRef]
  9. Nemeth, E.; Preza, G.C.; Jung, C.L.; Kaplan, J.; Waring, A.J.; Ganz, T. The N-terminus of hepcidin is essential for its interaction with ferroportin: Structure-function study. Blood 2006, 107, 328–333. [Google Scholar] [CrossRef]
  10. Theurl, I.; Theurl, M.; Seifert, M.; Mair, S.; Nairz, M.; Rumpold, H.; Zoller, H.; Bellmann-Weiler, R.; Niederegger, H.; Talasz, H.; et al. Autocrine formation of hepcidin induces iron retention in human monocytes. Blood 2008, 111, 2392–2399. [Google Scholar] [CrossRef] [Green Version]
  11. Liu, X.-B.; Nguyen, N.-B.H.; Marquess, K.D.; Yang, F.; Haile, D.J. Regulation of hepcidin and ferroportin expression by lipopolysaccharide in splenic macrophages. Blood Cells. Mol. Dis. 2005, 35, 47–56. [Google Scholar] [CrossRef]
  12. Hintze, K.J.; Snow, D.; Nabor, D.; Timbimboo, H. Adipocyte hypoxia increases hepatocyte hepcidin expression. Biol. Trace Elem. Res. 2011, 143, 764–771. [Google Scholar] [CrossRef]
  13. Kulaksiz, H.; Theilig, F.; Bachmann, S.; Gehrke, S.G.; Rost, D.; Janetzko, A.; Cetin, Y.; Stremmel, W. The iron-regulatory peptide hormone hepcidin: Expression and cellular localization in the mammalian kidney. J. Endocrinol. 2005, 184, 361–370. [Google Scholar] [CrossRef] [PubMed]
  14. Tabbah, S.M.; Buhimschi, C.S.; Rodewald-Millen, K.; Pierson, C.R.; Bhandari, V.; Samuels, P.; Buhimschi, I.A. Hepcidin, an Iron Regulatory Hormone of Innate Immunity, is Differentially Expressed in Premature Fetuses with Early-Onset Neonatal Sepsis. Am. J. Perinatol. 2018, 35, 865–872. [Google Scholar] [CrossRef] [PubMed]
  15. Gnana-Prakasam, J.P.; Martin, P.M.; Mysona, B.A.; Roon, P.; Smith, S.B.; Ganapathy, V. Hepcidin expression in mouse retina and its regulation via lipopolysaccharide/Toll-like receptor-4 pathway independent of Hfe. Biochem. J. 2008, 411, 79–88. [Google Scholar] [CrossRef]
  16. Lakhal-Littleton, S. Cardiomyocyte hepcidin: From intracellular iron homeostasis to physiological function. Vitam. Horm. 2019, 110, 189–200. [Google Scholar] [CrossRef] [PubMed]
  17. Kautz, L.; Meynard, D.; Monnier, A.; Darnaud, V.; Bouvet, R.; Wang, R.-H.; Deng, C.; Vaulont, S.; Mosser, J.; Coppin, H.; et al. Iron regulates phosphorylation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1, and Atoh8 in the mouse liver. Blood 2008, 112, 1503–1509. [Google Scholar] [CrossRef]
  18. Nicolas, G.; Chauvet, C.; Viatte, L.; Danan, J.L.; Bigard, X.; Devaux, I.; Beaumont, C.; Kahn, A.; Vaulont, S. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J. Clin. Investig. 2002, 110, 1037–1044. [Google Scholar] [CrossRef]
  19. Nemeth, E.; Rivera, S.; Gabayan, V.; Keller, C.; Taudorf, S.; Pedersen, B.K.; Ganz, T. IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin. J. Clin. Investig. 2004, 113, 1271–1276. [Google Scholar] [CrossRef]
  20. Pak, M.; Lopez, M.A.; Gabayan, V.; Ganz, T.; Rivera, S. Suppression of hepcidin during anemia requires erythropoietic activity. Blood 2006, 108, 3730–3735. [Google Scholar] [CrossRef] [Green Version]
  21. Arezes, J.; Foy, N.; McHugh, K.; Sawant, A.; Quinkert, D.; Terraube, V.; Brinth, A.; Tam, M.; LaVallie, E.R.; Taylor, S.; et al. Erythroferrone inhibits the induction of hepcidin by BMP6. Blood 2018, 132, 1473–1477. [Google Scholar] [CrossRef] [PubMed]
  22. Wang, C.Y.; Xu, Y.; Traeger, L.; Dogan, D.Y.; Xiao, X.; Steinbicker, A.U.; Babitt, J.L. Erythroferrone lowers hepcidin by sequestering BMP2/6 heterodimer from binding to the BMP type i receptor ALK3. Blood 2020, 135, 453–456. [Google Scholar] [CrossRef] [PubMed]
  23. Camaschella, C.; Nai, A.; Silvestri, L. Iron metabolism and iron disorders revisited in the hepcidin era. Haematologica 2020, 105, 260–272. [Google Scholar] [CrossRef]
  24. Drakesmith, H.; Nemeth, E.; Ganz, T. Ironing out Ferroportin. Cell Metab. 2015, 22, 777–787. [Google Scholar] [CrossRef]
  25. Billesbølle, C.B.; Azumaya, C.M.; Kretsch, R.C.; Powers, A.S.; Gonen, S.; Schneider, S.; Arvedson, T.; Dror, R.O.; Cheng, Y.; Manglik, A. Structure of hepcidin-bound ferroportin reveals iron homeostatic mechanisms. Nature 2020, 586, 807–811. [Google Scholar] [CrossRef] [PubMed]
  26. Aschemeyer, S.; Qiao, B.; Stefanova, D.; Valore, E.V.; Sek, A.C.; Alex Ruwe, T.; Vieth, K.R.; Jung, G.; Casu, C.; Rivella, S.; et al. Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Blood 2018, 131, 899–910. [Google Scholar] [CrossRef]
  27. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.D.; Donovan, A.; Ward, D.M.V.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef]
  28. Beguin, Y.; Stray, S.M.; Cazzola, M.; Huebers, H.A.; Finch, C.A. Ferrokinetic measurement of erythropoiesis. Acta Haematol. 1988, 79, 121–126. [Google Scholar] [CrossRef]
  29. Camaschella, C.; Nai, A. Ineffective erythropoiesis and regulation of iron status in iron loading anaemias. Br. J. Haematol. 2016, 172, 512–523. [Google Scholar] [CrossRef]
  30. Talawy, T.S.; Altoum, A.E.A.; Babker, A.M. The Role of Erythroferrone Hormone as Erythroid Regulator of Hepcidin and Iron Metabolism during Thalassemia and in Iron Deficiency Anemia- A Short Review. J. Pharm. Res. Int. 2020, 32, 55–59. [Google Scholar] [CrossRef]
  31. Kautz, L.; Jung, G.; Du, X.; Gabayan, V.; Chapman, J.; Nasoff, M.; Nemeth, E.; Ganz, T. Erythroferrone contributes to hepcidin suppression and iron overload in a mouse model of β-thalassemia. Blood 2015, 126, 2031–2037. [Google Scholar] [CrossRef] [PubMed]
  32. Kautz, L.; Jung, G.; Valore, E.V.; Rivella, S.; Nemeth, E.; Ganz, T. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat. Genet. 2014, 46, 678–684. [Google Scholar] [CrossRef] [PubMed]
  33. Srole, D.N.; Ganz, T. Erythroferrone structure, function, and physiology: Iron homeostasis and beyond. J. Cell. Physiol. 2021, 236, 4888–4901. [Google Scholar] [CrossRef] [PubMed]
  34. Finberg, K.E.; Heeney, M.M.; Campagna, D.R.; Aydinok, Y.; Pearson, H.A.; Hartman, K.R.; Mayo, M.M.; Samuel, S.M.; Strouse, J.J.; Markianos, K.; et al. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat. Genet. 2008, 40, 569–571. [Google Scholar] [CrossRef] [PubMed]
  35. Du, X.; She, E.; Gelbart, T.; Truksa, J.; Lee, P.; Xia, Y.; Khovananth, K.; Mudd, S.; Mann, N.; Moresco, E.M.Y.; et al. The serine protease TMPRSS6 is required to sense iron deficiency. Science 2008, 320, 1088–1092. [Google Scholar] [CrossRef] [PubMed]
  36. Camaschella, C.; Pagani, A.; Nai, A.; Silvestri, L. The mutual control of Iron and erythropoiesis. Int. J. Lab. Hematol. 2016, 38 (Suppl. 1), 20–26. [Google Scholar] [CrossRef] [PubMed]
  37. Brewin, J.; El Hoss, S.; Strouboulis, J.; Rees, D. A novel index to evaluate ineffective erythropoiesis in hematological diseases offers insights into sickle cell disease. Haematologica 2022, 107, 338–341. [Google Scholar] [CrossRef] [PubMed]
  38. Li, H.; Choesang, T.; Bao, W.; Chen, H.; Feola, M.; Garcia-Santos, D.; Li, J.; Sun, S.; Follenzi, A.; Pham, P.; et al. Decreasing TfR1 expression reverses anemia and hepcidin suppression in β-thalassemic mice. Blood 2017, 129, 1514–1526. [Google Scholar] [CrossRef]
  39. Forejtnikovà, H.; Vieillevoye, M.; Zermati, Y.; Lambert, M.; Pellegrino, R.M.; Guihard, S.; Gaudry, M.; Camaschella, C.; Lacombe, C.; Roetto, A.; et al. Transferrin receptor 2 is a component of the erythropoietin receptor complex and is required for efficient erythropoiesis. Blood 2010, 116, 5357–5367. [Google Scholar] [CrossRef]
  40. Artuso, I.; Lidonnici, M.R.; Altamura, S.; Mandelli, G.; Pettinato, M.; Muckenthaler, M.U.; Silvestri, L.; Ferrari, G.; Camaschella, C.; Nai, A. Transferrin receptor 2 is a potential novel therapeutic target for β-thalassemia: Evidence from a murine model. Blood 2018, 132, 2286–2297. [Google Scholar] [CrossRef] [Green Version]
  41. Origa, R.; Galanello, R.; Ganz, T.; Giagu, N.; Maccioni, L.; Faa, G.; Nemeth, E. Liver iron concentrations and urinary hepcidin in β-thalassemia. Haematologica 2007, 92, 583–588. [Google Scholar] [CrossRef] [PubMed]
  42. Kearney, S.L.; Nemeth, E.; Neufeld, E.J.; Thapa, D.; Ganz, T.; Weinstein, D.A.; Cunningham, M.J. Urinary hepcidin in congenital chronic anemias. Pediatr. Blood Cancer 2007, 48, 57–63. [Google Scholar] [CrossRef] [PubMed]
  43. Pasricha, S.-R.; McHugh, K.; Drakesmith, H. Regulation of Hepcidin by Erythropoiesis: The Story So Far. Annu. Rev. Nutr. 2016, 36, 417–434. [Google Scholar] [CrossRef] [PubMed]
  44. Jenkins, Z.A.; Hagar, W.; Bowlus, C.L.; Johansson, H.E.; Harmatz, P.; Vichinsky, E.P.; Theil, E.C. Iron homeostasis during transfusional iron overload in beta-thalassemia and sickle cell disease: Changes in iron regulatory protein, hepcidin, and ferritin expression. Pediatr. Hematol. Oncol. 2007, 24, 237–243. [Google Scholar] [CrossRef]
  45. Schmidt, P.J.; Fleming, M.D. Modulation of hepcidin as therapy for primary and secondary iron overload disorders: Preclinical models and approaches. Hematol. Oncol. Clin. N. Am. 2014, 28, 387–401. [Google Scholar] [CrossRef]
  46. Gardenghi, S.; Ramos, P.; Marongiu, M.F.; Melchiori, L.; Breda, L.; Guy, E.; Muirhead, K.; Rao, N.; Roy, C.N.; Andrews, N.C.; et al. Hepcidin as a therapeutic tool to limit iron overload and improve anemia in β-thalassemic mice. J. Clin. Investig. 2010, 120, 4466–4477. [Google Scholar] [CrossRef] [PubMed]
  47. Nicholls, A.; Likliter, J.; Tozzi, L.; Liu, D.; Shames, R. Hepcidin mimetic PTG-300 induces dose-related and sustained reductions in serum iron and transferrin saturation in healthy. In Proceedings of the 23rd Congress of the European Hematology Association, Stockholm, Sweden, 14–17 June 2018; p. S895. [Google Scholar]
  48. Bourne, G.; Li, Z.; Brandari, A.; Frederick, B.; McMahon, J.; Tran, V. Hepcidin mimetic PTG-300 for treatment of ineffective erythropoiesis and chronic anemia in hemoglobinopathy diseases. In Proceedings of the 23rd Congress of the European Hematology Association, Stockholm, Sweden, 14–17 June 2018; p. S843. [Google Scholar]
  49. Lal, A.; Voskaridou, E.; Flevari, P.; Taher, A.; Chew, L.-P.; Valone, F.; Gupta, S.; Viprakasit, V. A hepcidin mimetic, PTG-300, demonstrates pharmacodynamic effects indicating reduced iron availability in transfusion-dependent beta-thalassemia subjects. In Proceedings of the 25th European Hematology Association Annual Congress, Tokyo, Japan, 15 May 2020; p. S298. [Google Scholar]
  50. Casu, C.; Oikonomidou, P.R.; Chen, H.; Nandi, V.; Ginzburg, Y.; Prasad, P.; Fleming, R.E.; Shah, Y.M.; Valore, E.V.; Nemeth, E.; et al. Minihepcidin peptides as disease modifiers in mice affected by β-thalassemia and polycythemia vera. Blood 2016, 128, 265–276. [Google Scholar] [CrossRef]
  51. Casu, C.; Chessa, R.; Liu, A.; Gupta, R.; Drakesmith, H.; Fleming, R.; Ginzburg, Y.Z.; MacDonald, B.; Rivella, S. Minihepcidins improve ineffective erythropoiesis and splenomegaly in a new mouse model of adult β-thalassemia major. Haematologica 2020, 105, 1835–1844. [Google Scholar] [CrossRef]
  52. Monteiro, D.G.; van Dijk, J.W.A.; Aliyanto, R.; Fung, E.; Nemeth, E.; Ganz, T.; Rosengren, J.; Clark, R.J. Pursuing orally bioavailable hepcidin analogues via cyclic N-methylated mini-hepcidins. Biomedicines 2021, 9, 164. [Google Scholar] [CrossRef]
  53. Nai, A.; Pagani, A.; Mandelli, G.; Lidonnici, M.R.; Silvestri, L.; Ferrari, G.; Camaschella, C. Deletion of TMPRSS6 attenuates the phenotype in a mouse model of β-thalassemia. Blood 2012, 119, 5021–5029. [Google Scholar] [CrossRef]
  54. Guo, S.; Casu, C.; Gardenghi, S.; Booten, S.; Aghajan, M.; Peralta, R.; Watt, A.; Freier, S.; Monia, B.P.; Rivella, S. Reducing TMPRSS6 ameliorates hemochromatosis and β-thalassemia in mice. J. Clin. Investig. 2013, 123, 1531–1541. [Google Scholar] [CrossRef] [PubMed]
  55. Schmidt, P.J.; Toudjarska, I.; Sendamarai, A.K.; Racie, T.; Milstein, S.; Bettencourt, B.R.; Hettinger, J.; Bumcrot, D.; Fleming, M.D. An RNAi therapeutic targeting Tmprss6 decreases iron overload in Hfe(-/-) mice and ameliorates anemia and iron overload in murine β-thalassemia intermedia. Blood 2013, 121, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  56. Porter, J.; Taher, A.; Viprakasit, V.; Kattamis, A.; Coates, T.D.; Garbowski, M.; Dürrenberger, F.; Manolova, V.; Richard, F.; Cappellini, M.D. Oral ferroportin inhibitor vamifeport for improving iron homeostasis and erythropoiesis in β-thalassemia: Current evidence and future clinical development. Expert Rev. Hematol. 2021, 14, 633–644. [Google Scholar] [CrossRef]
  57. Richard, F.; van Lier, J.J.; Roubert, B.; Haboubi, T.; Göhring, U.M.; Dürrenberger, F. Oral ferroportin inhibitor VIT-2763: First-in-human, phase 1 study in healthy volunteers. Am. J. Hematol. 2020, 95, 68–77. [Google Scholar] [CrossRef]
  58. Arezes, J.; Foy, N.; McHugh, K.; Quinkert, D.; Benard, S.; Sawant, A.; Frost, J.N.; Armitage, A.E.; Pasricha, S.R.; Lim, P.J.; et al. Antibodies against the erythroferrone N-terminal domain prevent hepcidin suppression and ameliorate murine thalassemia. Blood 2020, 135, 547–557. [Google Scholar] [CrossRef] [PubMed]
  59. Casu, C.; Pettinato, M.; Liu, A.; Aghajan, M.; Lo Presti, V.; Lidonnici, M.R.; Munoz, K.A.; Hara, E.O.; Olivari, V.; Maria, S.; et al. Regular Article Correcting b -thalassemia by combined therapies that restrict Iron and modulate erythropoietin activity. Blood 2020, 136, 1968–1979. [Google Scholar] [CrossRef] [PubMed]
  60. Garbowski, M.W.; Evans, P.; Vlachodimitropoulou, E.; Hider, R.; Porter, J.B. Residual erythropoiesis protects against myocardial hemosiderosis in transfusiondependent thalassemia by lowering labile plasma iron via transient generation of apotransferrin. Haematologica 2017, 102, 1640–1649. [Google Scholar] [CrossRef] [PubMed]
  61. Casu, C.; Aghajan, M.; Oikonomidou, P.R.; Guo, S.; Monia, B.P.; Rivella, S. Combination of Tmprss6- ASO and the iron chelator deferiprone improves erythropoiesis and reduces iron overload in a mouse model of beta-thalassemia intermedia. Haematologica 2016, 101, e8–e11. [Google Scholar] [CrossRef]
  62. Schmidt, P.J.; Racie, T.; Westerman, M.; Fitzgerald, K.; Butler, J.S.; Fleming, M.D. Combination therapy with a Tmprss6 RNAi-therapeutic and the oral iron chelator deferiprone additively diminishes secondary iron overload in a mouse model of β-thalassemia intermedia. Am. J. Hematol. 2015, 90, 310–313. [Google Scholar] [CrossRef]
  63. Vadolas, J.; Ng, G.Z.; Kysenius, K.; Crouch, P.J.; Dames, S.; Eisermann, M.; Nualkaew, T.; Vilcassim, S.; Schaeper, U.; Grigoriadis, G. SLN124, a GalNac-siRNA targeting transmembrane serine protease 6, in combination with deferiprone therapy reduces ineffective erythropoiesis and hepatic iron-overload in a mouse model of β-thalassaemia. Br. J. Haematol. 2021. [Google Scholar] [CrossRef]
  64. Schmidt, P.J.; Fitzgerald, K.; Butler, J.S.; Fleming, M.D. Global loss of Tfr2 with concomitant induced iron deficiency greatly ameliorates the phenotype of a murine thalassemia intermedia model. Am. J. Hematol. 2021, 96, 251–257. [Google Scholar] [CrossRef] [PubMed]
  65. Nyffenegger, N.; Flace, A.; Doucerain, C.; Dürrenberger, F.; Manolova, V. The oral ferroportin inhibitor VIT-2763 improves erythropoiesis without interfering with iron chelation therapy in a mouse model of β-thalassemia. Int. J. Mol. Sci. 2021, 22, 873. [Google Scholar] [CrossRef] [PubMed]
  66. Lucarelli, G.; Galimberti, M.; Polchi, P.; Giardini, C.; Politi, P.; Baronciani, D.; Angelucci, E.; Manenti, F.; Delfini, C.; Aureli, G. Marrow transplantation in patients with advanced thalassemia. N. Engl. J. Med. 1987, 316, 1050–1055. [Google Scholar] [CrossRef] [PubMed]
  67. Baronciani, D.; Angelucci, E.; Potschger, U.; Gaziev, J.; Yesilipek, A.; Zecca, M.; Orofino, M.G.; Giardini, C.; Al-Ahmari, A.; Marktel, S.; et al. Hemopoietic stem cell transplantation in thalassemia: A report from the European Society for Blood and Bone Marrow Transplantation Hemoglobinopathy Registry, 2000–2010. Bone Marrow Transplant. 2016, 51, 536–541. [Google Scholar] [CrossRef] [PubMed]
  68. Nienhuis, A.W.; Persons, D.A. Development of gene therapy for thalassemia. Cold Spring Harb. Perspect. Med. 2012, 2, 1–17. [Google Scholar] [CrossRef]
  69. Schuessler-Lenz, M.; Enzmann, H.; Vamvakas, S. Regulators’ Advice Can Make a Difference: European Medicines Agency Approval of Zynteglo for Beta Thalassemia. Clin. Pharmacol. Ther. 2020, 107, 492–494. [Google Scholar] [CrossRef]
  70. Marktel, S.; Scaramuzza, S.; Cicalese, M.P.; Giglio, F.; Galimberti, S.; Lidonnici, M.R.; Calbi, V.; Assanelli, A.; Bernardo, M.E.; Rossi, C.; et al. Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent ß-thalassemia. Nat. Med. 2019, 25, 234–241. [Google Scholar] [CrossRef]
  71. Locatelli, F.; Thompson, A.A.; Kwiatkowski, J.L.; Porter, J.B.; Thrasher, A.J.; Hongeng, S.; Sauer, M.G.; Thuret, I.; Lal, A.; Algeri, M.; et al. Betibeglogene Autotemcel Gene Therapy for Non–β 0 /β 0 Genotype β-Thalassemia. N. Engl. J. Med. 2022, 386, 415–427. [Google Scholar] [CrossRef]
  72. Goyal, S.; Tisdale, J.; Schmidt, M.; Kanter, J.; Jaroscak, J.; Whitney, D.; Bitter, H.; Gregory, P.D.; Parsons, G.; Foos, M.; et al. Acute Myeloid Leukemia Case after Gene Therapy for Sickle Cell Disease. N. Engl. J. Med. 2022, 386, 138–147. [Google Scholar] [CrossRef]
  73. Jones, R.J.; DeBaun, M.R. Leukemia after gene therapy for sickle cell disease: Insertional mutagenesis, busulfan, both, or neither. Blood 2021, 138, 942–947. [Google Scholar] [CrossRef]
  74. Grace, R.F.; Rose, C.; Layton, D.M.; Galactéros, F.; Barcellini, W.; Morton, D.H.; van Beers, E.J.; Yaish, H.; Ravindranath, Y.; Kuo, K.H.M.; et al. Safety and Efficacy of Mitapivat in Pyruvate Kinase Deficiency. N. Engl. J. Med. 2019, 381, 933–944. [Google Scholar] [CrossRef]
  75. Matte, A.; Federti, E.; Kung, C.; Kosinski, P.A.; Narayanaswamy, R.; Russo, R.; Federico, G.; Carlomagno, F.; Desbats, M.A.; Salviati, L.; et al. The pyruvate kinase activator mitapivat reduces hemolysis and improves anemia in a β-thalassemia mouse model. J. Clin. Investig. 2021, 131, 1–12. [Google Scholar] [CrossRef] [PubMed]
  76. Kuo, K.H.M.; Layton, D.M.; Lal, A.; Al-Samkari, H.; Bhatia, J.; Kosinski, P.A.; Tong, B.; Lynch, M.; Uhlig, K.; Vichinsky, E.P. Safety and efficacy of mitapivat, an oral pyruvate kinase activator, in adults with non-transfusion dependent α-thalassaemia or β-thalassaemia: An open-label, multicentre, phase 2 study. Lancet 2022, 400, 493–501. [Google Scholar] [CrossRef]
  77. Forsyth, S.; Schroeder, P.; Geib, J.; Vrishabhendra, L.; Konstantinidis, D.G.; LaSalvia, K.; Ribadeneira, M.D.; Wu, E.; Kelly, P.; Kalfa, T.A. Safety, Pharmacokinetics, and Pharmacodynamics of Etavopivat (FT-4202), an Allosteric Activator of Pyruvate Kinase-R, in Healthy Adults: A Randomized, Placebo-Controlled, Double-Blind, First-in-Human Phase 1 Trial. Clin. Pharmacol. Drug Dev. 2022, 11, 654–665. [Google Scholar] [CrossRef]
  78. Piga, A.; Perrotta, S.; Gamberini, M.R.; Voskaridou, E.; Melpignano, A.; Filosa, A.; Caruso, V.; Pietrangelo, A.; Longo, F.; Tartaglione, I.; et al. Luspatercept improves hemoglobin levels and blood transfusion requirements in a study of patients with b-thalassemia. Blood 2019, 133, 1279–1289. [Google Scholar] [CrossRef] [PubMed]
  79. Cappellini, M.D.; Viprakasit, V.; Taher, A.T.; Georgiev, P.; Kuo, K.H.M.; Coates, T.; Voskaridou, E.; Liew, H.-K.; Pazgal-Kobrowski, I.; Forni, G.L.; et al. A Phase 3 Trial of Luspatercept in Patients with Transfusion-Dependent β-Thalassemia. N. Engl. J. Med. 2020, 382, 1219–1231. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Central role of ineffective erythropoiesis in hepcidin cascade in thalassemia (ERFE = erythroferrone; EPO = erythropoietin; ROS = reactive oxygen species).
Figure 1. Central role of ineffective erythropoiesis in hepcidin cascade in thalassemia (ERFE = erythroferrone; EPO = erythropoietin; ROS = reactive oxygen species).
Jcm 11 05119 g001
Figure 2. Potential new treatments emerging from advances in the pathophysiology of thalassemia (ERFE = erythroferrone; FPN = ferroportin; TMPRSS6 = transmembrane serine protease 6).
Figure 2. Potential new treatments emerging from advances in the pathophysiology of thalassemia (ERFE = erythroferrone; FPN = ferroportin; TMPRSS6 = transmembrane serine protease 6).
Jcm 11 05119 g002
Table 1. Treatments to increase hepcidin function in thalassemia (TD = transfusion-dependent; NTD = non-transfusion-dependent; ERFE = erythroferrone; FPN = ferroportin; HSCT = hematopoietic stem cell transplantation; TMPRSS6 = transmembrane serine protease 6; ASO = antisense oligonucleotide; siRNA = small interfering RNA; NTBI = non-transferrin-bound iron; TfR2 = transferin receptor 2; TfR1 = transferin receptor 1; BM = bone marrow).
Table 1. Treatments to increase hepcidin function in thalassemia (TD = transfusion-dependent; NTD = non-transfusion-dependent; ERFE = erythroferrone; FPN = ferroportin; HSCT = hematopoietic stem cell transplantation; TMPRSS6 = transmembrane serine protease 6; ASO = antisense oligonucleotide; siRNA = small interfering RNA; NTBI = non-transferrin-bound iron; TfR2 = transferin receptor 2; TfR1 = transferin receptor 1; BM = bone marrow).
ActionMechanismCompound/ModelThal TypeCTs?Clinicaltrials.gov
or Key Reference
Hepcidin mimeticHepcidin mimetics-Hepcidin LJPC-401
-Hepcidin PTG-300
-Hepcidin PTG-300
-Mini-hepcidin
TD
TD
TD/NTDT
YES
YES
YES
NO
-NCT03381833
-NCT03802201
-NCT04054921
-Goncalves, 2021
Hepcidin agonistTMPRSS6 inhibition-by RNA ASO
-by siRNA
-Ionis-TMPRSS6-LRx
-SLN124
NTD
NTD
YES
YES
-NCT04059406
-NCT04718844
FPN inhibitionVIT-2763NTDYES-NCT04364269
ERFE inhibitionN-Terminal ERFE Abs NOArezes, 2020
TfR2 inactivationBM TfR2KO mice NOArtuso, 2018
Apotransferrin↓ TfR1 expression
↓ NTBI
Human apotransferrinNTDYES-NCT03993613
Table 2. Potential combination of treatments targeting hepcidin function (TD = transfusion-dependent; NTD = non-transfusion-dependent; ERFE = erythroferrone; FPN = ferroportin; HSCT = hematopoietic stem cell transplantation; TMPRSS6 = transmembrane serine protease 6; ASO = antisense oligonucleotide; siRNA = small interfering RNA).
Table 2. Potential combination of treatments targeting hepcidin function (TD = transfusion-dependent; NTD = non-transfusion-dependent; ERFE = erythroferrone; FPN = ferroportin; HSCT = hematopoietic stem cell transplantation; TMPRSS6 = transmembrane serine protease 6; ASO = antisense oligonucleotide; siRNA = small interfering RNA).
Combination
12ConditionFindingsRef.
TMPRSS6 inhibitionIron chelation (Deferiprone)Hbbth3/+ mice↓ Liver ironVadolas J, 2021
TMPRSS6 inhibitionTfR2 inhibition
(Tfr2Y245X/Y245Xdouble mutant)
Hbbth3/+ mice↑ Hb
↓ Tissue iron
Schmidt PJ, 2020
TMPRSS6 inhibitionEPOHbbth3/+ mice↑ Hb
↓ splenomegaly
Casu C, 2020
TMPRSS6 inhibitionTfR2 single-allele deletionHbbth3/+ mice↑ Hb
↓ splenomegaly
Casu C, 2020
FPN inhibition Iron chelation (Deferasirox)Hbbth3/+ mice↑ Hb
↓ Liver iron
Nyffenegger N, 2021
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Longo, F.; Piga, A. Does Hepcidin Tuning Have a Role among Emerging Treatments for Thalassemia? J. Clin. Med. 2022, 11, 5119. https://doi.org/10.3390/jcm11175119

AMA Style

Longo F, Piga A. Does Hepcidin Tuning Have a Role among Emerging Treatments for Thalassemia? Journal of Clinical Medicine. 2022; 11(17):5119. https://doi.org/10.3390/jcm11175119

Chicago/Turabian Style

Longo, Filomena, and Antonio Piga. 2022. "Does Hepcidin Tuning Have a Role among Emerging Treatments for Thalassemia?" Journal of Clinical Medicine 11, no. 17: 5119. https://doi.org/10.3390/jcm11175119

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop