Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NICOTINAMIDE MONONUCLEOTIDE DERIVATIVES AND USE THEREOF IN THE STIMULATION OF THE EXPRESSION OF FETAL HEMOGLOBIN
Document Type and Number:
WIPO Patent Application WO/2022/129536
Kind Code:
A1
Abstract:
The present invention relates to nicotinamide mononucleotide derivatives of Formula (I) for use in the stimulation of the expression of fetal hemoglobin. The present invention further relates to pharmaceutical compositions comprising compounds of Formula (I) for use in the stimulation of the expression of fetal hemoglobin.

Inventors:
BERMOND GUILLAUME (FR)
GARCON LAURENT (FR)
CANAULT MATTHIAS (FR)
CROS CECILE (CH)
Application Number:
PCT/EP2021/086508
Publication Date:
June 23, 2022
Filing Date:
December 17, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NUVAMID SA (CH)
International Classes:
A61K31/662; A61K31/663; A61P7/06
Domestic Patent References:
WO2015186114A12015-12-10
WO2006105440A22006-10-05
WO2007008548A22007-01-18
WO2008128299A12008-10-30
Foreign References:
US4256108A1981-03-17
US4166452A1979-09-04
US4265874A1981-05-05
Other References:
PATON D M ET AL: "MECHANISM OF PRESYNAPTIC INHIBITION OF CHOLINERGIC TRANSMISSION IN GUINEA-PIG ILEUM BY ADENINE NUCLEOTIDES", METHODS AND FINDINGS IN EXPERIMENTAL AND CLINICAL PHARMACOLOGY, vol. 7, no. 2, 1985, pages 65 - 68, XP009527753, ISSN: 0379-0355
MOSCA A ET AL: "The relevance of hemoglobin F measurement in the diagnosis of thalassemias and related hemoglobinopathies", CLINICAL BIOCHEMISTRY, ELSEVIER, AMSTERDAM, NL, vol. 42, no. 18, 1 December 2009 (2009-12-01), pages 1797 - 1801, XP026782098, ISSN: 0009-9120, [retrieved on 20090704], DOI: 10.1016/J.CLINBIOCHEM.2009.06.023
KHALED M. MUSALLAMVIJAY G. SANKARANMARIA DOMENICA CAPPELLINILORENA DUCAL DAVID G. NATHANALI T. TAHER: "Fetal hemoglobin levels and morbidity in untransfused patients with 0-thalassemia intermedia", BLOOD, vol. 119, no. 2, 12 January 2012 (2012-01-12)
N.HERBERT ET AL.: "Individual red blood cell fetal hemoglobin quantification allows to determine protective thresholds in sickle cell disease", AM J HEMATOL., vol. 95, 2020, pages 1235 - 1245
WU LC ET AL., BLOOD, vol. 108, 2006, pages 1183 - 1188
Attorney, Agent or Firm:
AUGUST DEBOUZY (FR)
Download PDF:
Claims:
CLAIMS 1. A compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof; wherein: X is selected from O, CH2, S, Se, CHF, CF2 and C=CH2; R1 is selected from H, azido, cyano, (C1-C8)alkyl, (C1-C8) thio-alkyl, (C1-C8) heteroalkyl and OR; wherein R is selected from H and (C1-C8) alkyl; R2, R3, R4 and R5 are independently selected from H, halogen, azido, cyano, hydroxyl, (C1-C12) alkyl, (C1-C12)thioalkyl, (C1-C12)heteroalkyl, (C1-C12) haloalkyl and OR; wherein R is selected from H, (C1-C12) alkyl, -C(O)(C1-C12)alkyl, -C(O)NH(C1-C12)alkyl, -C(O)O(C1-C12)alkyl, -C(O)aryl, -C(O)(C1-C12)alkyl -(C5- C12)aryl, -C(O)NH(C1-C12)alkyl-C5-C12 aryl, -C(O)O(C1-C12)alkyl-(C5-C12)aryl and -C(O)CHRAANH2 ; wherein RAA is a side chain selected from a proteinogenic amino acid; R6 is selected from H, azido, cyano, (C1-C8) alkyl, (C1-C8) thioalkyl, (C1-C8) heteroalkyl and OR; wherein R is selected from H and (C1-C8) alkyl; R7 is selected from H, P(O)R9R10, P(S)R9R10 and  ; wherein: R9 and R10 are independently selected from OH, OR11, NHR13, NR13R14, (C1-C8) alkyl, (C2-C8) alkenyl, (C2-C8) alkynyl, (C3-C10) cycloalkyl, (C5-C12) aryl, (C5-C12) aryl-(C1-C8)alkyl, (C1-C8) alkyl-(C5-C12 aryl, (C1-C8) heteroalkyl, (C3-C8) heterocycloalkyl, (C5-C12) heteroaryl and NHCRαRα'C(O)OR12; wherein: - R11 is selected from (C1-C10 alkyl, (C3-C10) cycloalkyl, (C5-C12) aryl, (C1-C10 alkyl-(C5-C12) aryl, substituted (C5-C12) aryl, (C1-C10) heteroalkyl, (C1-C10 haloalkyl, -(CH2)mC(O)(C1-C15)alkyl, -(CH2)mOC(O)(C1-C15)alkyl, -(CH2)mOC(O)O(C1-C15)alkyl, -(CH2)mSC(O)(C1-C15)alkyl, -(CH2)mC(O)O(C1-C15)alkyl, -(CH2)mC(O)O(C1-C15)alkyl-(C5-C12)aryl; wherein m is an integer selected from 1 to 8; and -P(O)(OH)OP(O)(OH)2, , and an internal or external counterion; - R12 is selected from hydrogen, (C1-C10) alkyl, (C2-C8) alkenyl, (C2-C8) alkynyl, (C1-C10) haloalkyl, (C3-C10) cycloalkyl, (C3-C10)heterocycloalkyl, (C5-C12) aryl, (C1-C4) alkyl-(C5-C12) aryl and (C5-C12) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C1-C6) alkyl, (C1-C6) alkoxy and cyano; - R13 and R14 are independently selected from H, (C1-C8)alkyl and (C1-C8) alkyl-(C5-C12) aryl; - Rα and Rα’ are independently selected from an hydrogen, (C1-C10) alkyl, (C2-C10) alkenyl, (C2-C10) alkynyl, (C3-C10) cycloalkyl, (C1-C10) thioalkyl, (C1-C10 hydroxylalkyl, (C1-C10) alkyl-(C5-C12) aryl, (C5-C12) aryl, -(CH2)3NHC(=NH)NH2, (1H-indol-3-yl)methyl, (1H-imidazol-4-yl)methyl and a side chain selected from a proteinogenic or non-proteinogenic amino acid; wherein said aryl groups are optionally substituted with a group selected from hydroxyl, (C1-C10 )alkyl, (C1-C6) alkoxy, halogen, nitro and cyano; or R9 and R10 together with the phosphorus atom to which they are attached form a 6-membered ring wherein –R9−R10− represents –O-CH2-CH2-CHR-O−; wherein R is selected from hydrogen, (C5-C6) aryl and (C5-C6) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C1-C6) alkyl, (C1-C6) alkoxy and cyano; X’ is selected from O, CH2, S, Se, CHF, CF2 and C=CH2; R1’ is selected from H, azido, cyano, (C1-C8) alkyl, (C1-C8) thioalkyl, (C1-C8) heteroalkyl and OR; wherein R is selected from H and C1-C8 alkyl; R2’, R3’, R4’ and R5’ are independently selected from H, halogen, azido, cyano, hydroxyl, (C1-C12) alkyl, (C1-C12) thioalkyl, (C1-C12) heteroalkyl, (C1-C12) haloalkyl and OR; wherein R is selected from H, (C1-C12) alkyl, -C(O)(C1- C12)alkyl, -C(O)NH(C1-C12)alkyl, -C(O)O(C1-C12)alkyl, -C(O)aryl, -C(O)(C1-C12)alkyl-(C5-C12)aryl, -C(O)NH(C1-C12)alkyl-(C5-C12) aryl, -C(O)O(C1-C12)alkyl-(C5-C12) aryl and -C(O)CHRAANH2 ; wherein RAA is a side chain selected from a proteinogenic amino acid; R6’ is selected from H, azido, cyano, (C1-C8) alkyl, (C1-C8) thioalkyl, (C1-C8) heteroalkyl and OR; wherein R is selected from H and (C1-C8) alkyl; R8’ is selected from H, OR, NHR15’, NR15’R16’, NH-NHR15’, SH, CN, N3 and halogen; wherein R is selected from H and (C1-C8)alkyl, and R15’ and R16’ are independently selected from H, (C1-C8) alkyl and (C1-C8) alkyl-(C5-C12)aryl and -CHRAA’CO2H wherein RAA’ is a side chain selected from a proteinogenic or non- proteinogenic amino acid; Y’ is selected from CH, CH2, CHCH3, C(CH3)2 and CCH3; n is an integer selected from 1 to 3;  represents the point of attachment, represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R1’; R8 is selected from H, OR, NHR15, NR15R16, NH-NHR15, SH, CN, N3 and halogen; wherein R is selected from H and (C1-C8)alkyl, and R15 and R16 are independently selected from H, (C1-C8) alkyl and (C1-C8) alkyl-(C5-C12)aryl and -CHRAACO2H wherein RAA is a side chain selected from a proteinogenic or non-proteinogenic amino acid; Y is selected from CH, CH2, CHCH3, C(CH3)2 and CCH3; represents a single or double bond according to Y; and represents the alpha or beta anomer depending on the position of R1, for use in the stimulation of the expression of fetal hemoglobin. 2. The compound for use according to claim 1, wherein X represents an oxygen. 3. The compound for use according to claim 1 or claim 2, wherein R1 and R6 are identical and represent hydrogen. 4. The compound for use according to any one of claims 1 to 3, wherein R3 and R4 are identical and represent hydrogen. 5. The compound for use according to any one of claims 1 to 4, wherein R2 and R5 are identical and represent OH. 6. The compound for use according to any one of claims 1 to 5, wherein Y is selected from CH or CH2. 7. The compound for use according to any one of claims 1 to 6, wherein R7 is selected from H, P(O)R9R10 and ; wherein R9 and R10 are as described in claim 1 and wherein: X’ is an oxygen; R1’ and R6’ each represents a hydrogen; R2’, R3’, R4’ and R5’ are independently selected from hydrogen and OH; R8’ is NH2; Y’ is selected from CH and CH2; n is equal to 2; represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R1’. 8. The compound for use according to any one of claims 1 to 7, selected from: or pharmaceutically acceptable salts and solvates thereof.  9. The compound for use according to any one of claims 1 to 8, wherein the compound is selected from compounds 001, 002, 009, 010 and 011. 10. The compound for use according to any one of claims 1 to 9, wherein the compound is for use in a patient, preferably a patient having sickle cell disease or thalassemia. 11. The compound for use according to claim 10, wherein the patient is a child. 12. Pharmaceutical composition for use for the stimulation of the expression of fetal hemoglobin comprising at least one compound of formula (I) as defined in any one of claims 1 to 11 and at least one pharmaceutically acceptable carrier.

13. The pharmaceutical composition for use according to claim 12, comprising in addition to the at least one compound of formula (I) as defined in any one of claims 1 to 11, at least one other active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ (Pyrroloquinoline quinone); hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. 14. The pharmaceutical composition for use according to claim 12 or 13 for use in a patient, preferably a patient having sickle cell disease or thalassemia. 15. The pharmaceutical composition for use according to claim 14 wherein the patient is a child.

Description:
NICOTINAMIDE MONONUCLEOTIDE DERIVATIVES AND USE THEREOF IN THE STIMULATION OF THE EXPRESSION OF FETAL HEMOGLOBIN FIELD OF INVENTION The present invention relates to nicotinamide mononucleotide derivatives compounds for use in the stimulation of the expression of fetal hemoglobin, preferably in a patient having sickle cell disease or thalassemia, pharmaceuticals compositions comprising the same as well as methods of use of nicotinamide mononucleotide derivatives compounds. BACKGROUND OF INVENTION Sickle cell disease (SCD) or drepanocytosis is a group of inherited red blood cell disorders defined by a missense point mutation in the sequence of beta globin, which results in a glutamic acid residue at position 6 being substituted by a valine. This mutated globin, called sickle hemoglobin or hemoglobin S (HbS), aggregates, and forms fibrous precipitates upon low oxygen level, leading to polymerized hemoglobin and promoting red blood cell (RBC) sickling. Clinical manifestations of SCD derive from at least three different pathophysiologic mechanisms: the loss of deformability of the RBC leading to vascular obstruction and ischemia; a shortened lifespan of the RBC leading to both intravascular and extravascular hemolysis; a sticky RBC surface increasing adherence to the vascular endothelium which can result in vascular obstruction and can contribute to vascular proliferative lesions. Recurrent acute pain crises, or vaso-occlusive crises (VOCs) are considered among the most common manifestations of SCD. VOCs are believed to occur when blood flow is obstructed, usually at the level of the small blood vessels resulting in ischemic injury and pain. Over time patients will also experience significant acute and chronic complications. Acute complications include serious infections such as meningitis, osteomyelitis, and sepsis, and noninfectious complications such as stroke, renal necrosis, priapism. Acute chest syndrome is a potentially life-threatening complication that can involve chest pain and shortness of breath among other symptoms; some episodes of acute chest syndrome are triggered by infection. Chronic complications can emerge across multiple organs and include neurocognitive impairment, chronic kidney injury, delayed puberty, avascular necrosis, retinopathy, pulmonary hypertension, skin ulcers, and chronic pain. Individuals with SCD face ongoing and evolving lifelong difficulties as a result of their disease. SCD affects over 5 million subjects in the world, being the most common genetic disease in France. Despite the recent advances in the field, therapy for SCD patients is limited to symptomatic treatment of pain, oxygen supplementation, antibiotics, RBC transfusions and hydroxyurea. Nonetheless, blood transfusion remains the most applied therapy to treat patients suffering from SCD. Alternative approaches, such as bone marrow transplantation and gene therapy have been developed but are still associated with toxicity and are only considered in case of cerebral vasculopathy. Moreover, these approaches are not yet feasible in most countries where the incidence of the disease is elevated. Oxidative stress contributes to the complex pathophysiology of sickle cell disease. Nicotinamide adenine dinucleotide (NAD+) is a ubiquitous oxidation-reduction (redox) cofactor in RBCs. NAD+ and its reduced form, NADH, play major roles in maintaining redox balance. Sickle red cells have a lower redox ratio ([NADH]:[NAD++NADH]) than normal RBCs. The amino acid L-glutamine (USAN, glutamine) is required to synthesize NAD. Uptake of L-glutamine is several times greater in sickle red cells than in normal RBCs, primarily to increase the total intracellular NAD level. Oral administration of pharmaceutical-grade L-glutamine was shown to raise the NAD redox ratio within sickle cells and was associated with patient-reported clinical improvement. A phase 3 trial of L-glutamine in SCD showed that the median number of pain crises over 48 weeks was lower among the patients who received L-glutamine. On the basis of the results of this phase 3 trial, the FDA granted approval of pharmaceutical grade L- glutamine (Endari, Emmaus Medical) as a prescription drug to reduce the rate of acute complications of SCD among adults and children 5 years of age and older. Some other protocols have recently been granted by the FDA to treat SCD or reduce complications associated with SCD: Voxelotor (Oxbryta™), which inhibits polymerization of HbS by promoting the binding of oxygen to hemoglobin, has been approved to treat SCD in adults and children 12 years of age and older; and Crizanlizumab (Adakveo™), a therapeutic monoclonal antibody that reduces the phenomenon of cell aggregation during VOCs by inhibiting P-Selectin, a cell adhesion molecule, has been approved in adults and children 16 years of age and older. However, while current treatments have greatly increased the life expectancy of affected patients, they are still limited as the effectiveness of these drugs varies depending on the patient and the clinical manifestation observed. Moreover, further studies need to be done to assess whether the beneficial effect observed on SCD complications is preserved over the years. It has also been found that the RBCs from infants with SCD take longer to sickle and do not deform as much as their parents' suffering from SCD. Thalassemia syndromes result from deficiencies in either alpha-globin (alpha- thalassemia) or beta-like globin (beta-thalassemia) chains. The diseases become apparent when the deficient globin is required during development. Alpha-Thalassemia is symptomatic during gestation, as alpha-globin is required for fetal hemoglobin (HbF, alpha2-gamma2). As beta-globin is not required in large amounts before birth, beta- thalassemia is asymptomatic until around 6 months after birth. Mutations that cause prolonged production of fetal gamma-globin chains may present later, at 2 to 4 years of age. Fetal hemoglobin is found in fetal red blood cells and is the main oxygen carrier protein in the human fetus. Fetal hemoglobin can be noted HbF or α 2 γ 2 (alpha 2 -gamma 2 ). Fetal hemoglobin differs from the adult hemoglobin in that it comprises γ (gamma) subunits instead of β (beta) subunits. Fetal hemoglobin is produced at around 6 weeks of pregnancy and remains high until 2-4 months old of the born baby. The level of fetal hemoglobin decreases within the first year after birth and remains below 1% of total hemoglobin in children and adults. If fetal hemoglobin remains relatively high after birth, the number of painful episodes decreases in patients with SCD and they have a better prognosis. Fetal hemoglobin's role in reducing disease severity comes from its ability to disrupt the formation of HbS chains within RBCs. For these reasons, hydroxyurea has been proposed as a treatment for SCD by increasing HbF in RBCs of patients. However, hydroxyurea has many side effects such as nausea, vomiting, rash, pain, fast heartbeat, leg wounds and ulcers, increase of infections and bleedings, etc. It has also been demonstrated that high HbF levels are associated with a milder disease course in patients with beta-thalassemia intermedia, although the mechanism is not yet understood (Khaled M. Musallam, Vijay G. Sankaran, Maria Domenica Cappellini, Lorena Duca,1 David G. Nathan, and Ali T. Taher, Fetal hemoglobin levels and morbidity in untransfused patients with β-thalassemia intermedia, Blood, 12 January 2012, Volume 119, Number 2). The Applicant surprisingly found that the nicotinamide mononucleotide derivatives according to the invention are potent agents to increase the level of fetal hemoglobin in blood, especially in patients suffering from sickle cell disease. SUMMARY This invention thus relates to a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof; wherein: X is selected from O, CH 2 , S, Se, CHF, CF 2 and C= CH 2 ; R 1 is selected from H, azido, cyano, (C 1 -C 8 )alkyl, (C 1 -C 8 ) thio-alkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 2 , R 3 , R 4 and R 5 are independently selected from H, halogen, azido, cyano, hydroxyl, (C 1 -C 12 ) alkyl, (C 1 -C 12 )thioalkyl, (C 1 -C 12 )heteroalkyl, (C 1 -C 12 ) haloalkyl and OR; wherein R is selected from H, (C 1 -C 12 ) alkyl, -C(O)(C 1 -C 12 )alkyl, -C(O)NH(C 1 -C 12 )alkyl, -C(O)O(C 1 -C 12 )alkyl, -C(O)aryl, -C(O)(C 1 -C 12 )alkyl -(C 5 - C 12 )aryl, -C(O)NH(C 1 -C 12 )alkyl-C 5 -C 12 aryl, -C(O)O(C 1 -C 12 )alkyl-(C 5 -C 12 )aryl and -C(O)CHR AA NH 2 ; wherein R AA is a side chain selected from a proteinogenic amino acid; R 6 is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thioalkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 7 is selected from H, P(O)R 9 −R 10 P(S)R 9 R 10 and  ; wherein: R 9 and R 10 are independently selected from OH, OR 11 , NHR 13 , NR 13 R 14 , (C 1 -C 8 ) alkyl, (C 2 -C 8 ) alkenyl, (C 2 -C 8 ) alkynyl, (C 3- C 10 ) cycloalkyl, (C 5 -C 12 ) aryl, (C 5 -C 12 ) aryl-(C 1 -C 8 )alkyl, (C 1 -C 8 ) alkyl-(C 5 -C 12 aryl, (C 1 -C 8 ) heteroalkyl, (C 3 -C 8 ) heterocycloalkyl, (C 5 -C 12 ) heteroaryl and NHCRαRα'C(O)OR 12 ; wherein: - R 11 is selected from (C 1 -C 10 alkyl, (C 3 -C 10 ) cycloalkyl, (C 5 -C 12 ) aryl, (C 1 -C 10 alkyl-(C 5 -C 12 ) aryl, substituted (C 5 -C 12 ) aryl, (C 1 -C 10 ) heteroalkyl, (C 1 -C 10 haloalkyl, -(CH 2 ) m C(O)(C 1 -C 15 )alkyl, -(CH 2 ) m OC(O)(C 1 -C 15 )alkyl, -(CH 2 ) m OC(O)O(C 1 -C 15 )alkyl, -(CH 2 ) m SC(O)(C 1 -C 15 )alkyl, -(CH 2 ) m C(O)O(C 1 -C 15 )alkyl, -(CH 2 ) m C(O)O(C 1 -C 15 )alkyl-(C 5 -C 12 )aryl; wherein m is an integer selected from 1 to 8; and -P(O)(OH)OP(O)(OH) 2, , and an internal or external counterion; - R 12 is selected from hydrogen, (C 1 -C 10 ) alkyl, (C 2 -C 8 ) alkenyl, (C 2 -C 8 ) alkynyl, (C 1- C 10 ) haloalkyl, (C 3- C 10 ) cycloalkyl, (C 3- C 10 )heterocycloalkyl, (C 5 -C 12 ) aryl, (C 1 -C 4 ) alkyl-(C 5 -C 12 ) aryl and (C 5 -C 12 ) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy and cyano; - R 13 and R 14 are independently selected from H, (C 1 -C 8 )alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 aryl; - R α and R α' are independently selected from an hydrogen, (C 1- C 10 ) alkyl, (C 2 -C 10 ) alkenyl, (C 2 -C 10 ) alkynyl, (C 3 -C 10 ) cycloalkyl, (C 1 -C 10 ) thioalkyl, (C 1 -C 10 hydroxylalkyl, (C 1 -C 10 ) alkyl-(C 5 -C 12 ) aryl, (C 5 -C 12 ) aryl, -(CH 2 ) 3 NHC(=NH)NH 2 , (1H-indol-3-yl)methyl, (1H-imidazol-4-yl)methyl and a side chain selected from a proteinogenic or non-proteinogenic amino acid; wherein said aryl groups are optionally substituted with a group selected from hydroxyl, (C 1- C 10 )alkyl, (C 1- C 6 ) alkoxy, halogen, nitro and cyano; or R 9 and R 10 together with the phosphorus atom to which they are attached form a 6-membered ring wherein –R 9 −R 10 − represents –O-CH 2 -CH 2 -CHR-O−; wherein R is selected from hydrogen, (C 5 -C 6 ) aryl and (C 5 -C 6 ) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy and cyano; X’ is selected from O, CH 2 , S, Se, CHF, CF 2 and C=CH 2 ; R 1’ is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thio-alkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and C 1 -C 8 alkyl; R 2’ , R 3’ , R 4’ and R 5’ are independently selected from H, halogen, azido, cyano, hydroxyl, (C 1 -C 12 ) alkyl, (C 1 -C 12 ) thioalkyl, (C 1 -C 12 ) heteroalkyl, (C 1 -C 12 ) haloalkyl and OR; wherein R is selected from H, (C 1 -C 12 ) alkyl, -C(O)(C 1 - C 12 )alkyl, -C(O)NH(C 1 -C 12 )alkyl, -C(O)O(C 1 -C 12 )alkyl, -C(O)aryl, -C(O)(C 1 -C 12 )alkyl-(C 5 -C 12 )aryl, -C(O)NH(C 1 -C 12 )alkyl-(C 5 -C 12 ) aryl, -C(O)O(C 1 -C 12 )alkyl-(C 5 -C 12 ) aryl and -C(O)CHR AA NH 2 ; wherein R AA is a side chain selected from a proteinogenic amino acid; R 6’ is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thioalkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 8’ is selected from H, OR, NHR 15' , NR 15' R 16’ , NH-NHR 15' , SH, CN, N 3 and halogen; wherein R is selected from H and (C 1 -C 8 )alkyl, and R 15’ and R 16’ are independently selected from H, (C 1 -C 8 ) alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 )aryl and -CHR AA' CO 2 H wherein R AA' is a side chain selected from a proteinogenic or non- proteinogenic amino acid; Y’ is selected from CH, CH 2 , CHCH 3 , C(CH 3 ) 2 and CCH 3 ; n is an integer selected from 1 to 3;  represents the point of attachment, represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R 1’ ; R 8 is selected from H, OR, NHR 15 , NR 15 R 16 , NH-NHR 15 , SH, CN, N 3 and halogen; wherein R is selected from H and (C 1 -C 8 )alkyl, and R 15 and R 16 are independently selected from H, (C 1 -C 8 ) alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 )aryl and -CHR AA CO 2 H wherein R AA is a side chain selected from a proteinogenic or non-proteinogenic amino acid; Y is selected from CH, CH 2 , CHCH 3 , C(CH 3 ) 2 and CCH 3 ; represents a single or double bond according to Y; and represents the alpha or beta anomer depending on the position of R 1 , for use in the stimulation of the expression of fetal hemoglobin. According to one embodiment, X represents an oxygen. According to one embodiment, R 1 and R 6 are identical and represent hydrogen. According to one embodiment, R 3 and R 4 are identical and represent hydrogen. According to one embodiment, R 2 and R 5 are identical and represent OH. According to one embodiment, R 8 is NH 2 . According to one embodiment, Y is selected from CH and CH 2 . According to one embodiment, R 11 can be an internal or external counterion. According to one embodiment, wherein R 7 is selected from H, P(O)R 9 R 10 or ; wherein R 9 and R 10 are as described above and wherein: X’ is an oxygen; R 1’ and R 6’ each represents a hydrogen; R 2’ , R 3’ , R 4’ and R 5’ are independently selected from hydrogen or OH; R 8’ is NH 2 ; Y’ is selected from CH or CH 2; n is equal to 2;  represents the point of attachment, represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R 1’ .  According to one embodiment, the compound according to the invention is selected from:

or pharmaceutically acceptable salts and solvates thereof.   According to one embodiment, the compound according to the invention is selected from compounds 001, 002, 009, 010 and 011. According to an embodiment, the compounds of the invention increase the amount of fetal hemoglobin in the blood of a subject and/or the number of RBCs expressing fetal hemoglobin. According to an embodiment, the amount of fetal globin in the blood of the subject increases. According to an embodiment, the percentage of fetal globin in the blood of the subject increases. According to an embodiment, the number of F-cells in the blood of the subject increases. According to an embodiment, the amount of total hemoglobin in the blood of the subject increases. According to an embodiment, the number of F- reticulocytes in the blood of the subject increases. According to one embodiment, the compounds of the invention are for use in combination with at least one other active ingredient from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ (Pyrroloquinoline quinone); hydroxyurea, L- glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. According to one embodiment, the compounds of the invention are for use in a subject. According to one embodiment, said subject is a human. According to one embodiment, said subject is a human patient having sickle cell disease or a thalassemia. According to one embodiment, said subject is a human patient having beta-thalassemia. According to one embodiment, said subject is a human patient having alpha-thalassemia. According to one embodiment, said subject is a child. According to one embodiment, said child is 10 or young than 10 years old, preferably 5 or young than 5 years old, more preferably 2 or younger than 2 years old. According to one embodiment, said subject is a child having sickle cell disease. According to one embodiment, said subject is a child having a thalassemia, preferably alpha-thalassemia or beta-thalassemia. According to one embodiment, said subject is a child having beta-thalassemia. The present invention further relates to a pharmaceutical composition for the stimulation of the expression of fetal hemoglobin comprising at least one compound of formula (I) as defined herein above and at least one pharmaceutically acceptable carrier. According to one embodiment, the pharmaceutical composition for use according to the invention, comprises in addition to the at least one compound of formula (I) as defined herein above, at least one other active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ (Pyrroloquinoline quinone); hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. According to one embodiment, said subject is a human. According to one embodiment, said subject is a human patient having sickle cell disease or a thalassemia. According to one embodiment, said subject is a human patient having beta-thalassemia. According to one embodiment, said subject is a human patient having alpha-thalassemia. According to one embodiment, said subject is a child. According to one embodiment, said child is 10 or young than 10 years old, preferably 5 or young than 5 years old, more preferably 2 or younger than 2 years old. According to one embodiment, said subject is a child having sickle cell disease. According to one embodiment, said subject is a child having a thalassemia, preferably alpha-thalassemia or beta-thalassemia. According to one embodiment, said subject is a child having beta-thalassemia. According to one embodiment, said composition is adapted for oral administration. The present invention further relates to a method for increasing the expression of fetal hemoglobin comprising the administration of compounds of formula (I) or a pharmaceutically acceptable salt or solvate thereof as defined above. According to an embodiment, said administration is by oral administration. According to an embodiment, said oral administration is by a gastro-resistant capsule or gastro-resistant tablet. According to one embodiment, said method increases the amount of fetal hemoglobin in the blood of a subject and/or the number RBCs expressing fetal hemoglobin. According to one embodiment, said subject is a human. According to one embodiment, said subject is a human patient having sickle cell disease or a thalassemia. According to one embodiment, said subject is a human patient having beta-thalassemia. According to one embodiment, said subject is a human patient having alpha-thalassemia. According to one embodiment, said subject is a child. According to one embodiment, said child is 10 or young than 10 years old, preferably 5 or young than 5 years old, more preferably 2 or younger than 2 years old. According to one embodiment, said subject is a child having sickle cell disease. According to one embodiment, said subject is a child having a thalassemia, preferably alpha-thalassemia or beta-thalassemia. According to one embodiment, said subject is a child having beta-thalassemia. According to an embodiment, said method comprises the further administration of at least one other active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ (Pyrroloquinoline quinone); hydroxyurea, L- glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. DEFINITIONS The definitions and explanations below are for the terms as used throughout the entire application, including both the specification and the claims. When describing the compounds of the invention, the terms used are to be construed in accordance with the following definitions, unless indicated otherwise. Unless indicated otherwise, the nomenclature of substituents that are not explicitly defined herein are arrived at by naming the adjacent functionality toward the point of attachment followed by the terminal portion of the functionality. For example, the substituent "arylalkyl" refers to the group -(aryl)-(alkyl). In the present invention, the following terms have the following meanings: The term "alkyl" by itself or as part of another substituent refers to a hydrocarbyl radical of Formula C n H 2n+1 wherein n is a number greater than or equal to 1. Generally, alkyl groups of this invention comprise from 1 to 12 carbon atoms, preferably from 1 to 10 carbon atoms, preferably from 1 to 8 carbon atoms, more preferably from 1 to 6 carbon atoms, still more preferably 1 to 2 carbon atoms. Alkyl groups may be linear or branche. Suitable alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, s-butyl and t-butyl, pentyl and its isomers (e.g. n-pentyl, iso-pentyl), hexyl and its isomers (e.g. n-hexyl, isohexyl), heptyl and its isomers (e.g. n-heptyl, iso-heptyl), octyl and its isomers (e.g. n-octyl, iso-octyl), nonyl and its isomers (e.g. n-nonyl, iso-nonyl), decyl and its isomers (e.g. n-decyl, iso-decyl), undecyl and its isomers, dodecyl and its isomers. Preferred alkyl groups include methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, s-butyl, t-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl and n-decyl. Saturated branched alkyls include, without being limited to, i-propyl, s-butyl, i-butyl, t-butyl, i-pentyl, 2-methylbutyl, 3-methylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylbutyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,3-dimethylhexyl, 2,4-dimethylhexyl, 2,5-dimethylhexyl, 2,2-dimethylpentyl, 2,2-dimethylhexyl, 3,3-dimethylpentyl, 3,3-dimethylhexyl, 4,4-dimethylhexyl, 2-ethylpentyl, 3-ethylpentyl, 2-ethylhexyl, 3-ethylhexyl, 4-ethylhexyl, 2-methyl-2-ethylpentyl, 2-methyl-3-ethylpentyl, 2-methyl-4-ethylpentyl, 2-methyl-2-ethylhexyl, 2-methyl-3-ethylhexyl, 2-methyl-4-ethylhexyl, 2,2-diethylpentyl, 3,3-diethylhexyl, 2,2-diethylhexyl, 3,3-diethylhexyl. Cx-Cy-alkyl refers to alkyl groups which comprise x to y carbon atoms. When the suffix "ene" ("alkylene") is used in conjunction with an alkyl group, this is intended to mean the alkyl group as defined herein having two single bonds as points of attachment to other groups. The term "alkylene" includes methylene, ethylene, methylmethylene, propylene, ethylethylene, and 1,2- dimethylethylene. The term "alkenyl" as used herein refers to an unsaturated hydrocarbyl group, which may be linear or branched, comprising one or more carbon-carbon double bonds. Suitable alkenyl groups comprise between 2 and 12 carbon atoms, preferably between 2 and 8 carbon atoms, still more preferably between 2 and 6 carbon atoms. Examples of alkenyl groups are ethenyl, 2- propenyl, 2-butenyl, 3-butenyl, 2-pentenyl and its isomers, 2-hexenyl and its isomers, 2,4-pentadienyl and the like. The term "alkynyl" as used herein refers to a class of monovalent unsaturated hydrocarbyl groups, wherein the unsaturation arises from the presence of one or more carbon-carbon triple bonds. Alkynyl groups typically, and preferably, have the same number of carbon atoms as described above in relation to alkenyl groups. Non limiting examples of alkynyl groups are ethynyl, 2- propynyl, 2-butynyl, 3-butynyl, 2-pentynyl and its isomers, 2-hexynyl and its isomers-and the like. The term "alkoxy" as used herein refers to any group –O-alkyl, wherein alkyl is as defined above. Suitable alkoxy groups include for example methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, t-butoxy, sec-butoxy, and n-pentoxy. The term "amino acid" as used herein refers to an alpha-aminated carboxylic acid, i.e. a molecule comprising a carboxylic acid functional group and an amine functional group in alpha position of the carboxylic acid group, for example a proteinogenic amino acid or a non-proteinogenic amino acid. The term "aryl" as used herein refers to a polyunsaturated, aromatic hydrocarbyl group having a single ring (i.e. phenyl) or multiple aromatic rings fused together (e.g. naphtyl) or linked covalently, typically containing 5 to 12 atoms; preferably 6 to 10, wherein at least one ring is aromatic. The aromatic ring may optionally include one to two additional rings (either cycloalkyl, heterocyclyl or heteroaryl) fused thereto. Aryl is also intended to include the partially hydrogenated derivatives of the carbocyclic systems enumerated herein. Non-limiting examples of aryl comprise phenyl, biphenylyl, biphenylenyl, 5- or 6-tetralinyl, naphthalen-1- or -2-yl, 4-, 5-, 6 or 7-indenyl, 1- 2-, 3-, 4- or 5- acenaphtylenyl, 3-, 4- or 5-acenaphtenyl, 1- or 2-pentalenyl, 4- or 5-indanyl, 5-, 6- , 7- or 8-tetrahydronaphthyl, 1,2,3,4-tetrahydronaphthyl, 1,4-dihydronaphthyl, 1-, 2-, 3-, 4- or 5-pyrenyl. The term "cycloalkyl" as used herein is a cyclic alkyl group, alkenyl or alkynyl that is to say, a monovalent, saturated, or unsaturated hydrocarbyl group having 1 or 2 cyclic structures. Cycloalkyl includes monocyclic or bicyclic hydrocarbyl groups. Cycloalkyl groups may comprise 3 or more carbon atoms in the ring and generally, according to this invention comprise from 3 to 10, more preferably from 3 to 8 carbon atoms still more preferably from 3 to 6 carbon atoms. Examples of cycloalkyl groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, with cyclopropyl being particularly preferred. The term "halo" or "halogen" means fluoro, chloro, bromo, or iodo. Preferred halo groups are fluoro and chloro. The term "haloalkyl" alone or as part of another group, refers to an alkyl radical having the meaning as defined above wherein one or more hydrogen atoms are replaced with a halogen as defined above. Non-limiting examples of such haloalkyl radicals include chloromethyl, 1-bromoethyl, fluoromethyl, difluoromethyl, trifluoro methyl, 1,1,1- trifluoroethyl and the like. C x -C y -haloalkyl are alkyl groups which comprise x to y carbon atoms. Preferred haloalkyl groups are difluoromethyl and trifluoromethyl. The term "heteroalkyl" means an alkyl group as defined above in which one or more carbon atoms are replaced by a heteroatom selected from oxygen, nitrogen and sulfur atoms. In heteroalkyl groups, the heteroatoms are linked along the alkyl chain only to carbon atoms, i.e. each heteroatom is separated from any other heteroatom by at least one carbon atom. However, the nitrogen and sulphur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. A heteroalkyl is bonded to another group or molecule only through a carbon atom, i.e. the bonding atom is not selected from the heteroatoms included in the heteroalkyl group. Where at least one carbon atom in an aryl group is replaced with a heteroatom, the resultant ring is referred to herein as a heteroaryl ring. The term "heteroaryl" as used herein by itself or as part of another group refers but is not limited to 5 to 12 carbon-atom aromatic rings or ring systems containing 1 to 2 rings which are fused together or linked covalently, typically containing 5 to 6 atoms; at least one of which is aromatic, in which one or more carbon atoms in one or more of these rings is replaced by oxygen, nitrogen and/or sulfur atoms where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Such rings may be fused to an aryl, cycloalkyl, heteroaryl or heterocyclyl ring. Non-limiting examples of such heteroaryl, include: furanyl, thiophenyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, oxatriazolyl, thiatriazolyl, pyridinyl, pyrimidyl, pyrazinyl, pyridazinyl, oxazinyl, dioxinyl, thiazinyl, triazinyl, imidazo[2,1-b][1,3] thiazolyl, thieno [3,2-b] furanyl, thieno [3,2-b]thiophenyl, thieno[2,3-d][1,3]thiazolyl, thieno[2,3-d]imidazolyl, tetrazolo[l,5-a]pyridinyl, indolyl, indolizinyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, isobenzothiophenyl, indazolyl, benzimidazolyl, 1,3-benzoxazolyl, 1,2-benzisoxazolyl, 2,1-benzisoxazolyl, 1,3-benzothiazolyl, 1,2-benzoisothiazolyl, 2,1-benzoisothiazolyl, benzotriazolyl, 1,2,3-benzoxadiazolyl, 2,1,3-benzoxadiazolyl, 1,2,3-benzothiadiazolyl, 2,1,3-benzothiadiazolyl, thienopyridinyl, purinyl, imidazo[1,2-a]pyridinyl, 6-oxo-pyridazin-1(6H)-yl, 2-oxopyridin-1(2H)-yl, 6-oxo-pyridazin-1(6H)-yl, 2-oxopyridin-1(2H)-yl, 1,3-benzodioxolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, quinoxalinyl. Where at least one carbon atom in a cycloalkyl group is replaced with a heteroatom, the resultant ring is referred to herein as "heterocycloalkyl" or "heterocyclyl". The terms "heterocyclyl", "heterocycloalkyl" or "heterocyclo" as used herein by itself or as part of another group refer to non-aromatic, fully saturated or partially unsaturated cyclic groups (for example, 3 to 7 member monocyclic, 7 to 11 member bicyclic, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Any of the carbon atoms of the heterocyclic group may be substituted by oxo (for example piperidone, pyrrolidinone). The heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows. The rings of multi- ring heterocycles may be fused, bridged and/or joined through one or more spiro atoms. Non limiting exemplary heterocyclic groups include oxetanyl, piperidinyl, azetidinyl, 2-imidazolinyl, pyrazolidinyl imidazolidinyl, isoxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, 3H- indolyl, indolinyl, isoindolinyl, 2-oxopiperazinyl, piperazinyl, homopiperazinyl, 2-pyrazolinyl, 3-pyrazolinyl, tetrahydro-2H-pyranyl, 2H-pyranyl, 4H-pyranyl, 3,4-dihydro-2H-pyranyl, 3-dioxolanyl, 1,4-dioxanyl, 2,5-dioximidazolidinyl, 2- oxopiperidinyl, 2-oxopyrrolodinyl, tetrahydropyranyl, tetrahydrofuranyl, tetrahydroquinolinyl, tetrahydroisoquinolin- 1-yl, tetrahydroisoquinolin-2-yl, tetrahydroisoquinolin-3-yl, tetrahydroisoquinolin-4-yl, thiomorpholin-4-yl, thiomorpholin-4-ylsulf oxide, thiomorpholin-4-ylsulfone, 1,3- dioxolanyl, 1,4-oxathianyl, 1H-pyrrolizinyl, tetrahydro-1,1-dioxothiophenyl, N- formylpiperazinyl, and morpholin-4-yl. The term "hydroxyalkyl" refers to an alkyl radical having the meaning as defined above wherein one or more hydrogen atoms are replaced with -OH moieties. The term "thio-alkyl" refers to an alkyl radical having the meaning as defined above wherein one or more hydrogen atoms are replaced with -SH moieties. The term “non-proteinogenic amino acid” as used herein refers to an amino acid not naturally encoded or found in the genetic code of living organism. Non limiting examples of non-proteinogenic amino acid are ornithine, citrulline, argininosuccinate, homoserine, homocysteine, cysteine-sulfinic acid, 2-aminomuconic acid, δ-aminolevulinic acid, β-alanine, cystathionine, γ-aminobutyrate, DOPA, 5-hydroxytryptophan, D-serine, ibotenic acid, α-aminobutyrate, 2-aminoisobutyrate, D-leucine, D-valine, D-alanine or D-glutamate . The term "proteinogenic amino acid" as used herein refers to an amino acid that is incorporated into proteins during translation of messenger RNA by ribosomes in living organisms, i.e. Alanine (ALA), Arginine (ARG), Asparagine (ASN), Aspartate (ASP), Cysteine (CYS), Glutamate (glutamic acid) (GLU), Glutamine (GLN), Glycine (GLY), Histidine (HIS), Isoleucine (ILE), Leucine (LEU), Lysine (LYS), Methionine (MET), Phenylalanine (PHE), Proline (PRO), Pyrrolysine (PYL), Selenocysteine (SEL), Serine (SER), Threonine (THR), Tryptophan (TRP), Tyrosine (TYR) or Valine (VAL). The term "prodrug" as used herein means the pharmacologically acceptable derivatives of compounds of Formula (I) such as esters whose in vivo biotransformation product is the active drug. Prodrugs are characterized by increased bio-availability and are readily metabolized into the active compounds in vivo. Suitable prodrugs for the purpose of the invention include phosphoramidates, HepDirect, (S)-acyl-2-thioethyl (SATE), carboxylic esters, in particular alkyl esters, aryl esters, acyloxyalkyl esters, and dioxolene carboxylic esters; ascorbic acid esters. The term "substituent" or "substituted" means that a hydrogen radical on a compound or group is replaced by any desired group which is substantially stable under the reaction conditions in an unprotected form or when protected by a protecting group. Examples of preferred substituents include, without being limited to, halogen (chloro, iodo, bromo, or fluoro); alkyl; alkenyl; alkynyl, as described above; hydroxy; alkoxy; nitro; thiol; thioether; imine; cyano; amido; phosphonato; phosphine; carboxyl; thiocarbonyl; sulfonyl; sulfonamide; ketone; aldehyde; ester; oxygen (-O); haloalkyl (e.g., trifluoromethyl); cycloalkyl, which may be monocyclic or fused or non-fused polycyclic (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or a heterocycloalkyl, which may be monocyclic or fused or non-fused polycyclic (e.g., pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiazinyl), monocyclic or fused or non-fused polycyclic aryl or heteroaryl (e.g., phenyl, naphthyl, pyrrolyl, indolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridyl, quinolinyl, isoquinolinyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, benzimidazolyl, benzothiophenyl, or benzofuranyl); amino (primary, secondary, or tertiary); CO 2 CH 3 ; CONH 2 ; OCH 2 CONH 2 ; NH 2 ; SO 2 NH 2 ; OCHF 2 ; CF 3 ; OCF 3 ; and such moieties may also be optionally substituted by a fused-ring structure or bridge, for example -OCH 2 O-. These substituents may optionally be further substituted with a substituent selected from such groups. In certain embodiments, the term "substituent" or the adjective "substituted" refers to a substituent selected from the group consisting of an alkyl, an alkenyl, an alkynyl, an cycloalkyl, an cycloalkenyl, a heterocycloalkyl, an aryl, a heteroaryl, an arylalkyl, a heteroarylalkyl, a haloalkyl, -C(O)NR 17 R 18 , -NR 19 C(O)R20, a halo, -OR 19 , cyano, nitro, a haloalkoxy, -C(O)R 19 , -NR 17 R 18 , -SR 19 , -C(O)OR 19 , -OC(O)R 19 , -NR 19 C(O)NR 17 R 18 , -OC(O)NR 17 R 18 , -NR 19 C(O)OR 20 , -S(O)rR 19 , -NR 19 S(O)Rr 20 , -OS(O)Rr 20 , S(O)rNR 17 R 18 , -O, -S, and -N-R 19 , wherein r is 1 or 2; R 17 and R 18 , for each occurrence are, independently, H, an optionally substituted alkyl, an optionally substituted alkenyl, an optionally substituted alkynyl, an optionally substituted cycloalkyl, an optionally substituted cycloalkenyl, an optionally substituted heterocycloalkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted arylalkyl, or an optionally substituted heteroarylalkyl; or R 17 and R 18 taken together with the nitrogen to which they are attached is optionally substituted heterocycloalkyl or optionally substituted heteroaryl; and R 19 and R 20 for each occurrence are, independently, H, an optionally substituted alkyl, an optionally substituted alkenyl, an optionally substituted alkynyl, an optionally substituted cycloalkyl, an optionally substituted cycloalkenyl, an optionally substituted heterocycloalkyl, an optionally substituted aryl, an optionally substituted heteroaryl, an optionally substituted arylalkyl, or an optionally substituted heteroarylalkyl. In certain embodiments, the term "substituent" or the adjective "substituted" refers to a solubilizing group. The bonds of an asymmetric carbon can be represented here using a solid triangle , a dashed triangle or a zigzag line . The term “active ingredient” refers to a molecule or a substance whose administration to a subject slows down or stops the progression, aggravation, or deterioration of one or more symptoms of a disease, or condition; alleviates the symptoms of a disease or condition; cures a disease or condition. According to one embodiment, the therapeutic ingredient is a small molecule, either natural or synthetic. According to another embodiment, the therapeutic ingredient is a biological molecule such as for example an oligonucleotide, a siRNA, a miRNA, a DNA fragment, an aptamer, an antibody and the like. The term "administration", or a variant thereof (e.g., “administering"), means providing the active agent or active ingredient, alone or as part of a pharmaceutically acceptable composition, to the patient in whom/which the condition, symptom, or disease is to be treated. The term “drug” refers to any substance that causes a change in physiology or psychology of a subject when administrated to the subject. In the context of the invention, “drug” encompasses both drugs for medical use (“medicinal drug” or “active ingredient”) and drugs for non-medical use, e.g., recreational drugs (e.g., psychoactive drugs). By "pharmaceutically acceptable" is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the patient. The term “pharmaceutically acceptable excipient”, “pharmaceutically acceptable carrier” or “pharmaceutical vehicle” refers to an inert medium or carrier used as a solvent or diluent in which the pharmaceutically active ingredient is formulated and/or administered, and which does not produce an adverse, allergic or other reaction when administered to an animal, preferably a human being. This includes all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic agents, absorption retardants and other similar ingredients. For human administration, preparations must meet standards of sterility, general safety and purity as required by regulatory agencies such as the FDA or EMA. For the purposes of the invention, "pharmaceutically acceptable excipient" includes all pharmaceutically acceptable excipients as well as all pharmaceutically acceptable carriers, diluents, and/or adjuvants. The term “pharmaceutically acceptable salts” include the acid addition and base salts . Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminum, arginine, benzathine, calcium, choline, diethylamine, 2-(diethylamino)ethanol, diolamine, ethanolamine, glycine, 4-(2-hydroxyethyl)-morpholine, lysine, magnesium, meglumine, morpholine, olamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. Pharmaceutically acceptable salts of compounds of Formula (I) may be prepared by one or more of these methods: (i) by reacting the compound of Formula (I) with the desired acid; (ii) by reacting the compound of Formula (I) with the desired base; (iii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of Formula (I) or by ring-opening a suitable cyclic precursor, e.g., a lactone or lactam, using the desired acid; and/or (iv) by converting one salt of the compound of Formula (I) to another by reaction with an appropriate acid or by means of a suitable ion exchange column. All these reactions are typically carried out in solution. The salt may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the salt may vary from completely ionized to almost non-ionized. Although generally, with respect to the salts of the compounds of the invention, pharmaceutically acceptable salts are preferred, it should be noted that the invention in its broadest sense also includes non-pharmaceutically acceptable salts, which may for example be used in the isolation and/or purification of the compounds of the invention. For example, salts formed with optically active acids or bases may be used to form diastereoisomeric salts that can facilitate the separation of optically active isomers of the compounds of Formula (I). The term "solvate" is used herein to describe a molecular complex comprising a compound of the invention and containing stoichiometric or sub-stoichiometric amounts of one or more pharmaceutically acceptable solvent molecule, such as ethanol. The term 'hydrate' refers to a solvate when said solvent is water. The term "administration", or a variant thereof (e.g., “administering"), means providing the active agent or active ingredient, alone or as part of a pharmaceutically acceptable composition, to the patient in whom/which the condition, symptom, or disease is to be treated or prevented. The term "human" refers to a subject of both genders and at any stage of development (i.e., neonate, infant, juvenile, adolescent, adult). The term “child” refers to a human below 18 of age. The term "subject" refers to a mammal, preferably a human. According to the present invention, a subject is a mammal, preferably a human, suffering from a red blood cell disorder and/or one or more complications associated with a red blood cell disorder, especially sickle cell disease and/or complications associated with sickle cell disease. In one embodiment, the subject is a “patient”, i.e., a mammal, preferably a human, who/which is awaiting the receipt of, or is receiving medical care or was/is/will be the object of a medical procedure or is monitored for the development of a red blood cell disorder and/or one or more complications associated with a red blood cell disorder, especially sickle cell disease and/or one or more complications associated with sickle cell disease. The terms “treat”, “treating” or “treatment”, as used herein, refer to a therapeutic treatment, to a prophylactic (or preventative) treatment, or to both a therapeutic treatment and a prophylactic (or preventive) treatment, wherein the object is to prevent, reduce, alleviate, and/or slow down (lessen) one or more of the symptoms of a red blood cell disorder and/or of the complications associated with a red blood cell disorder, especially sickle cell disease and/or complications associated with sickle cell disease, in a subject in need thereof. In one embodiment, “treating” or “treatment” refers to a therapeutic treatment. In another embodiment, “treating” or “treatment” refers to a prophylactic or preventive treatment. In yet another embodiment, “treating” or “treatment” refers to both a prophylactic (or preventive) treatment and a therapeutic treatment. The terms “prevent”, “preventing” and “prevention”, as used herein, refer to a method of delaying or precluding the onset of a condition or disease and/or at least one symptom, barring a patient from acquiring a condition or disease, or reducing a patient’s risk of acquiring a condition or disease. The term “therapeutically effective amount” (or more simply an “effective amount”) as used herein means the amount of active agent or active ingredient that is aimed at, without causing significant negative or adverse side effects to the subject in need of stimulation of the expression of fetal hemoglobin. DETAILED DESCRIPTION The present invention thus relates to the use of nicotinamide mononucleotide derivatives for the stimulation of expression of fetal hemoglobin. Preferably, the invention relates to the use of nicotinamide mononucleotide derivatives for the stimulation of expression of fetal hemoglobin by red blood cells of a patient having sickle cell disease or thalassemia. Compound for use in the stimulation of the expression of fetal hemoglobin In one embodiment, the nicotinamide mononucleotide derivative of the present invention is a compound of Formula (I), or a pharmaceutically acceptable salt or solvate thereof; wherein: X is selected from O, CH 2 , S, Se, CHF, CF 2 and C=CH 2 ; R 1 is selected from H, azido, cyano, (C 1 -C 8 )alkyl, (C 1 -C 8 ) thio-alkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 2 , R 3 , R 4 and R 5 are independently selected from H, halogen, azido, cyano, hydroxyl, (C 1 -C 12 ) alkyl, (C 1 -C 12 )thioalkyl, (C 1 -C 12 )heteroalkyl, (C 1 -C 12 ) haloalkyl and OR; wherein R is selected from H, (C 1 -C 12 ) alkyl, -C(O)(C 1 -C 12 )alkyl, -C(O)NH(C 1 -C 12 )alkyl, -C(O)O(C 1 -C 12 )alkyl, -C(O)aryl, -C(O)(C 1 -C 12 )alkyl -(C 5 - C 12 )aryl, -C(O)NH(C 1 -C 12 )alkyl-C 5 -C 12 aryl, -C(O)O(C 1 -C 12 )alkyl-(C 5 -C 12 )aryl and -C(O)CHR AA NH 2 ; wherein R AA is a side chain selected from a proteinogenic amino acid; R 6 is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thioalkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 7 is selected from H, P(O)R 9 R 10 , P(S)R 9 R 10 and  ; wherein: R 9 and R 10 are independently selected from OH, OR 11 , NHR 13 , NR 13 R 14 , (C 1 -C 8 ) alkyl, (C 2 -C 8 ) alkenyl, (C 2 -C 8 ) alkynyl, (C 3- C 10 ) cycloalkyl, (C 5 -C 12 ) aryl, (C 5 -C 12 ) aryl-(C 1 -C 8 )alkyl, (C 1 -C 8 ) alkyl-(C 5 -C 12 aryl, (C 1 -C 8 ) heteroalkyl, (C 3- C 8 ) heterocycloalkyl, (C 5 -C 12 ) heteroaryl and NHCR α R α’ C(O)OR 12 ; wherein: - R 11 is selected from (C 1 -C 10 alkyl, (C 3 -C 10 ) cycloalkyl, (C 5 -C 12 ) aryl, (C 1 -C 10 alkyl-(C 5 -C 12 ) aryl, substituted (C 5 -C 12 ) aryl, (C 1 -C 10 ) heteroalkyl, (C 1 -C 10 haloalkyl, -(CH 2 ) m C(O)(C 1 -C 15 )alkyl, -(CH 2 ) m OC(O)(C 1 -C 15 )alkyl, -(CH 2 ) m OC(O)O(C 1 -C 15 )alkyl, -(CH 2 ) m SC(O)(C 1 -C 15 )alkyl, -(CH 2 ) m C(O)O(C 1 -C 15 )alkyl, -(CH 2 ) m C(O)O(C 1 -C 15 )alkyl-(C 5 -C 12 )aryl; wherein m is an integer selected from 1 to 8; and -P(O)(OH)OP(O)(OH) 2, , and an internal or external counterion; - R 12 is selected from hydrogen, (C 1 -C 10 ) alkyl, (C 2 -C 8 ) alkenyl, (C 2 -C 8 ) alkynyl, (C 1- C 10 ) haloalkyl, (C 3- C 10 ) cycloalkyl, (C 3- C 10 )heterocycloalkyl, (C 5 -C 12 ) aryl, (C 1 -C 4 ) alkyl-(C 5 -C 12 ) aryl and (C 5 -C 12 ) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy and cyano; - R 13 and R 14 are independently selected from H, (C 1 -C 8 )alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 aryl; - Rα and Rα’ are independently selected from an hydrogen, (C 1 -C 10 ) alkyl, (C 2 -C 10 ) alkenyl, (C 2 -C 10 ) alkynyl, (C 3 -C 10 ) cycloalkyl, (C 1 -C 10 ) thioalkyl, (C 1- C 10 hydroxylalkyl, (C 1 -C 10 ) alkyl-(C 5 -C 12 ) aryl, (C 5 -C 12 ) aryl, -(CH 2 ) 3 NHC(=NH)NH 2 , (1H-indol-3-yl)methyl, (1H-imidazol-4-yl)methyl and a side chain selected from a proteinogenic or non-proteinogenic amino acid; wherein said aryl groups are optionally substituted with a group selected from hydroxyl, (C 1- C 10 )alkyl, (C 1- C 6 ) alkoxy, halogen, nitro and cyano; or R 9 and R 10 together with the phosphorus atom to which they are attached form a 6-membered ring wherein –R 9 −R 10 − represents –O-CH 2 -CH 2 -CHR-O−; wherein R is selected from hydrogen, (C 5 -C 6 ) aryl and (C 5 -C 6 ) heteroaryl; wherein said aryl or heteroaryl groups are optionally substituted by one or two groups selected from halogen, trifluoromethyl, (C 1 -C 6 ) alkyl, (C 1 -C 6 ) alkoxy and cyano; X’ is selected from O, CH 2 , S, Se, CHF, CF 2 and C=CH 2 ; R 1’ is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thio-alkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and C 1 -C 8 alkyl; R 2’ , R 3’ , R 4’ and R 5’ are independently selected from H, halogen, azido, cyano, hydroxyl, (C 1 -C 12 ) alkyl, (C 1 -C 12 ) thioalkyl, (C 1 -C 12 ) heteroalkyl, (C 1 -C 12 ) haloalkyl and OR; wherein R is selected from H, (C 1 -C 12 ) alkyl, -C(O)(C 1 - C 12 )alkyl, -C(O)NH(C 1 -C 12 )alkyl, -C(O)O(C 1 -C 12 )alkyl, -C(O)aryl, -C(O)(C 1 -C 12 )alkyl-(C 5 -C 12 )aryl, -C(O)NH(C 1 -C 12 )alkyl-(C 5 -C 12 ) aryl, -C(O)O(C 1 -C 12 )alkyl-(C 5 -C 12 ) aryl and -C(O)CHR AA NH 2 ; wherein R AA is a side chain selected from a proteinogenic amino acid; R 6’ is selected from H, azido, cyano, (C 1 -C 8 ) alkyl, (C 1 -C 8 ) thioalkyl, (C 1 -C 8 ) heteroalkyl and OR; wherein R is selected from H and (C 1 -C 8 ) alkyl; R 8’ is selected from H, OR, NHR 15’ , NR 15’ R 16’ , NH-NHR 15’ , SH, CN, N 3 and halogen; wherein R is selected from H and (C 1 -C 8 )alkyl, and R 15’ and R 16’ are independently selected from H, (C 1 -C 8 ) alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 )aryl and -CHR AA' CO 2 H wherein R AA' is a side chain selected from a proteinogenic or non- proteinogenic amino acid; Y’ is selected from CH, CH 2 , CHCH 3 , C(CH 3 )2 and CCH 3 ; n is an integer selected from 1 to 3;  represents the point of attachment, represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R 1’ ; R 8 is selected from H, OR, NHR 15 , NR 15 R 16 , NH-NHR 15 , SH, CN, N 3 and halogen; wherein R is selected from H and (C 1 -C 8 )alkyl, and R 15 and R 16 are independently selected from H, (C 1 -C 8 ) alkyl and (C 1 -C 8 ) alkyl-(C 5 -C 12 )aryl and -CHR AA CO 2 H wherein R AA is a side chain selected from a proteinogenic or non-proteinogenic amino acid; Y is selected from CH, CH 2 , CHCH 3 , C(CH 3 ) 2 and CCH 3 ; represents a single or double bond according to Y; and represents the alpha or beta anomer depending on the position of R 1 , for use in the stimulation of the expression of fetal hemoglobin. Otherwise stated, the compounds of the invention increase the amount of fetal hemoglobin in the blood and/or the number of red blood cells expressing fetal hemoglobin in a subject in need thereof and/or the proportion (%) of red blood cells expressing fetal hemoglobin. The nicotinamide mononucleotide derivatives of the invention may comprise one or more charged atoms. Particularly, when present, the phosphate groups may bear one or more charge, preferably one or more negative charge. Moreover, the nitrogen atom of the pyridine part of the nicotinamide group may bear one positive charge when it is quaternized. The presence of one or more charged atom in the nicotinamide mononucleotide derivatives of the invention depends on the conditions, especially pH conditions, that one skilled in the art will recognize. According to one embodiment, X is selected from O, CH 2 and S. In one embodiment, X is oxygen. According to one embodiment, R 1 is selected from hydrogen and OH. In one embodiment, R 1 is hydrogen. In one embodiment, R 1 is OH. According to one embodiment, R 2 , R 3 , R 4 and R 5 are independently selected from hydrogen, halogen, hydroxyl, C 1 -C 12 alkyl and OR; wherein R is as described herein above. In a preferred embodiment, R 2 , R 3 , R 4 and R 5 are independently selected from hydrogen, hydroxyl and OR; wherein R is as described herein above. In a more preferred embodiment R 2 , R 3 , R 4 and R 5 are independently selected from hydrogen and OH. According to one embodiment, R 2 and R 3 are identical. In one embodiment, R 2 and R 3 are identical and represent OH. In one embodiment, R 2 and R 3 are identical and represent hydrogen. According to one embodiment, R 2 and R 3 are different. In a preferred embodiment, R 2 is hydrogen and R 3 is OH. In a more preferred embodiment, R 2 is OH and R 3 is hydrogen. According to one embodiment, R 4 and R 5 are identical. In one embodiment, R 4 and R 5 are identical and represent OH. In one embodiment, R 4 and R 5 are identical and represent hydrogen. According to one embodiment, R 4 and R 5 are different. In a preferred embodiment, R 4 is OH and R 5 is hydrogen. In a more preferred embodiment, R 4 is hydrogen and R 5 is OH. According to one embodiment, R 3 and R 4 are different. In one embodiment, R 3 is OH and R 4 is hydrogen. In one embodiment, R 3 is hydrogen and R 4 is OH. According to one embodiment, R 3 and R 4 are identical. In a preferred embodiment, R 3 and R 4 are identical and represent OH. In a more preferred embodiment, R 3 and R 4 are identical and represent hydrogen. According to one embodiment, R 2 and R 5 are different. In one embodiment, R 2 is hydrogen and R 5 is OH. In one embodiment, R 2 is OH and R 5 is hydrogen. According to one embodiment, R 2 and R 5 are identical. In a preferred embodiment, R 2 and R 5 are identical and represent hydrogen. In a more preferred embodiment, R 2 and R 5 are identical and represent OH. According to one embodiment, R 6 is selected from hydrogen and OH. In one embodiment, R 6 is OH. In a preferred embodiment, R 6 is hydrogen. According to one embodiment, R 1 and R 6 are each independently selected from hydrogen and OH. According to one embodiment, R 1 and R 6 are both hydrogen atoms. According to one embodiment, R 7 is selected from hydrogen, P(O)R 9 R 10 and According to one embodiment, R 7 is selected from hydrogen, P(O)R 9 R 10 and According to one embodiment, R 7 is selected from P(O)R 9 R 10 and According to one embodiment, R 7 is hydrogen. In another embodiment, R 7 is not a hydrogen atom. According to one embodiment, R 7 is P(O)R 9 R 10 ; wherein R 9 and R 10 are as described herein above. In a preferred embodiment, R 7 is P(O)(OH) 2 . According to another embodiment, R 7 is wherein R 1’ , R 2’ , R 3’ , R 4’ , R 5’ , R 6’ , R 8’ , R 9 , X’, Y’, n, and are as described herein above for compounds of Formula (I). According to a preffered embodiment, R is  7 ; wherein: X’ is selected from O, CH 2 and S, preferably X’ is O; R 1’ is selected from hydrogen and OH, preferably R 1’ is hydrogen; R 2’ , R 3’ , R 4’ and R 5’ are independently selected from hydrogen, halogen, hydroxyl, C 1 -C 12 alkyl and OR; wherein R is as described herein above, preferably R 2’ , R 3’ , R 4’ and R 5’ are independently selected from hydrogen, hydroxyl and OR; wherein R is as described herein above, more preferably R 2’ , R 3’ , R 4’ and R 5’ are independently selected from hydrogen and OH; R 6’ is selected from hydrogen and OH, preferably R 6’ is hydrogen; R 8’ is selected from H, OR, NHR 15' and NR 15' R 16 ’; wherein R, R 15' and R 16 ’ are as described herein above, preferably R 8’ is NHR 15' ; wherein R 15' is as described herein above, more preferably R 8’ is NH 2 ;  Y’ is selected from CH or CH 2 n is an integer selected from 1 to 3; represents a single or double bond according to Y’; and represents the alpha or beta anomer depending on the position of R 1’ . According to one embodiment, n is 1. According to one embodiment, n is 2. According to one embodiment, n is 3. According to one embodiment, R 8 is selected from H, OR, NHR 15 or NR 15 R 16; wherein R 15 and R 16 are as described herein above. In a preferred embodiment, R 8 is NHR 15 ; wherein R 15 is as described herein above. In a preferred embodiment, R 8 is NH 2 .   According to one embodiment, Y is a CH or CH 2 . In one embodiment, Y is a CH. In one embodiment, Y is a CH 2 . According to a preferred embodiment, the invention relates to compounds of general Formula (II): or a pharmaceutically acceptable salt or solvate thereof; wherein R 7 is H and wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , X, Y, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-1): or a pharmaceutically acceptable salt or solvate thereof; wherein X is hydrogen and wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-2): or a pharmaceutically acceptable salt or solvate thereof; wherein R 1 is hydrogen and wherein R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and   are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-3): or a pharmaceutically acceptable salt or solvate thereof; R 3 and R 4 are identical and represent hydrogen and wherein R 2 , R 5 , R 6 , R 8 , Y,, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-4): or a pharmaceutically acceptable salt or solvate thereof; wherein R 2 and R 5 are identical and represent OH and wherein R 6 , R 8 , Y, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-5): or a pharmaceutically acceptable salt or solvate thereof; wherein R 6 is hydrogen and wherein R 8 , Y, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-6): or a pharmaceutically acceptable salt or solvate thereof; wherein R 8 is NH 2 and wherein Y, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (II) are those of Formula (II-7): or a pharmaceutically acceptable salt or solvate thereof; wherein Y is CH and wherein  is as described herein above for compounds of Formula (I). According to a preferred embodiment, the invention relates to compounds of general Formula (II-8):

or a pharmaceutically acceptable salt or solvate thereof; wherein Y is CH 2 and wherein and are as described herein above for compounds of Formula (I). According to another preferred embodiment, the invention relates to compounds of general Formula (III): or a pharmaceutically acceptable salt or solvate thereof; wherein R 7 is P(O)(OH) 2 and wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , X, Y, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-1): or a pharmaceutically acceptable salt or solvate thereof; wherein X is hydrogen and wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-2): or a pharmaceutically acceptable salt or solvate thereof; wherein R 1 is hydrogen and wherein R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and   are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-3): or a pharmaceutically acceptable salt or solvate thereof; R 3 and R 4 are identical and represent hydrogen and wherein R 2 , R 5 , R 6 , R 8 , Y,, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-4): or a pharmaceutically acceptable salt or solvate thereof; wherein R 2 and R 5 are identical and represent OH and wherein R 6 , R 8 , Y, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-5): or a pharmaceutically acceptable salt or solvate thereof; wherein R 6 is hydrogen and wherein R 8 , Y, and   are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-6): or a pharmaceutically acceptable salt or solvate thereof; wherein R 8 is NH 2 and wherein Y , and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-7): or a pharmaceutically acceptable salt or solvate thereof; wherein Y is CH and wherein is as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (III) are those of Formula (III-8): or a pharmaceutically acceptable salt or solvate thereof; wherein Y is CH 2 and wherein and are as described herein above for compounds of Formula (I). According to another preferred embodiment, the invention relates to compounds of general Formula (IV): or a pharmaceutically acceptable salt or solvate thereof; wherein n is equal to 2 and wherein R 1 , R 1’ , R 2 , R 2’ , R 3 , R 3’ , R 4 , R 4’ , R 5 , R 5’ , R 6 , R 6’ , R 8 , R 8’ , X, X’, Y, Y’, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-1): or a pharmaceutically acceptable salt or solvate thereof; wherein X and X’ are an oxygen and wherein R 1 , R 1’ , R 2 , R 2’ , R 3 , R 3’ , R 4 , R 4’ , R 5 , R 5’ , R 6 , R 6’ , R 8 , R 8’ , Y, Y’, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-2): or a pharmaceutically acceptable salt or solvate thereof; wherein R 1 and R 1’ are hydrogen and wherein R 2 , R 2’ , R 3 , R 3’ , R 4 , R 4’ , R 5 , R 5’ , R 6 , R 6’ , R 8 , R 8’ , Y, Y’, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-3): or a pharmaceutically acceptable salt or solvate thereof; wherein R 3 , R 3’ , R 4 and R 4’ are identical and represent hydrogen and wherein R 2 , R 2’ , R 5 , R 5’ , R 6 , R 6’ , R 8 , R 8’ , Y, Y’, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-4):

or a pharmaceutically acceptable salt or solvate thereof; wherein R 2 , R 2’ , R 5 and R 5’ are identical and represent OH and wherein R 6 , R 6’ , R 8 , R 8’ , Y, Y’, and  are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-5): or a pharmaceutically acceptable salt or solvate thereof; wherein R 6 and R 6’ are hydrogen and wherein R 6 , R 6’ , R 8 , R 8’ , Y, Y’, and are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-6): or a pharmaceutically acceptable salt or solvate thereof; wherein R 8 and R 8’ are NH 2 and wherein Y, Y’, and   are as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-7): or a pharmaceutically acceptable salt or solvate thereof; wherein Y and Y’ are CH and wherein  is as described herein above for compounds of Formula (I). According to one embodiment, preferred compounds of general Formula (IV) are those of Formula (IV-8): or a pharmaceutically acceptable salt or solvate thereof; wherein Y and Y’ are CH 2 and wherein is as described herein above for compounds of Formula (I). According to one embodiment, the compound according to the invention is selected from compounds 001 to 014 from Table 1 below or a pharmaceutically acceptable salt or solvate thereof:

[Table 1] According to one embodiment, preferred compounds of the invention are compounds 001 to 014 or a pharmaceutically acceptable salt or solvate thereof. According to one embodiment, more preferred compounds of the invention are compounds 001, 002, 009, 010 and 011. According to one embodiment, more preferred compounds of the invention are compounds 001 and 002 or a pharmaceutically acceptable salt or solvate thereof. According to another embodiment, more preferred compounds of the invention are compounds 009, 010 and 011 or a pharmaceutically acceptable salt or solvate thereof. According to one embodiment, even more preferred compounds of the invention are compounds 002, 010 and 011 or a pharmaceutically acceptable salt or solvate thereof. All references to compounds of Formula (I) include references to salts, solvates, multi-component complexes and liquid crystals thereof. All references to compounds of Formula (I) include references to polymorphs and crystal habits thereof. All references to compounds of Formula (I) include references to pharmaceutically acceptable prodrugs and prodrugs thereof. Process According to another aspect, the invention relates to a method for the preparation of the compound of Formula (I) as described hereinabove. In particular, the compounds of Formula (I) may be prepared as described below from substrates A-E. It shall be understood by a person skilled in the art that these schemes are in no way limiting and that variations may be made without departing from the spirit and scope of this invention. According to one embodiment, the method involves in a first step the mono-phosphorylation of a compound of Formula (A), in the presence of phosphoryl chloride and a trialkyl phosphate, to yield the phosphorodichloridate of Formula (B): wherein X, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and are as described herein above. In a second step, the phosphorodichloridate of Formula (B) is hydrolyzed to yield the phosphate of Formula (C): wherein X, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , Y, and are as described herein above. In an alternative embodiment, when in Formula (I) R 7 is , the phosphate compound of Formula (C) obtained in the second step is then reacted, with a phosphorodichloridate compound of Formula (B’) obtained as described in the first step: wherein R 1’ , R 2’ , R 3’ , R 4’ , R 5’ , R 6’ , R 8’ , X’, Y’, and are as described herein above;to give the compound of Formula (I) as described herein above; followed by hydrolysis to yield tothe compound of Formula (I). According to one embodiment, the compound of Formula (A) is synthesized using various methods known to the person skilled in the art. According to one embodiment, the compound of Formula (A) wherein Y is CH, referred to as compound of Formula (A-a), is synthesized by reacting the pentose of Formula (D) with a nitrogen derivative of Formula (E) leading to the compound of Formula (A-1), which is then selectively deprotected to give the compound of Formula (A-a), wherein X, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 8 , Y, and are as described herein above and R is a protective group. According to one embodiment, R is an appropriate protective group known to the skilled person in the art. In one embodiment, the protecting group is selected from triarylmethyls and silyls. Non-limiting examples of triarylmethyl include trityl, monomethoxytrityl, 4,4'-dimethoxytrityl and 4,4',4"-trimethoxytrityl. Non-limiting examples of silyl groups include trimethylsilyl, tert-butyldimethylsilyl, triisopropylsilyl, tert-butyldiphenylsilyl, tri-iso-propylsilyloxymethyl and [2-(trimethylsilyl)ethoxy]methyl. According to one embodiment, any hydroxyl group attached to the pentose is protected by an appropriate protective group known to the person skilled in the art. The choice and exchange of protective groups is the responsibility of the person skilled in the art. Protective groups can also be removed by methods well known to the skilled person, for example, with an acid (e.g. mineral or organic acid), base or fluoride source. According to a preferred embodiment, the nitrogen nicotinamide of Formula (E) is coupled to the pentose of Formula (D) by a reaction in the presence of a Lewis acid leading to the compound of Formula (A-1). Non-limiting examples of Lewis acids include TMSOTf, BF 3 .OEt 2 , TiCl 4 and FeCl 3 . According to one embodiment, the method of the present invention further comprises a step of reducing the compound of Formula (A-a) by various methods well known to the skilled person in the art, leading to the compound of Formula (A-b) wherein Y is CH 2 and X, R 1 , R 2 , R3, R 4 , R 5 , R 6 , R 8 , and are as defined above. According to a specific embodiment, the present invention relates to a method for the preparation of the compounds 001, 003, 005, 007 and 009:. In a first step, the nicotinamide of Formula (E-i) is coupled to the ribose tetraacetate of Formula (D-i) by a coupling reaction in the presence of a Lewis acid, resulting in the compound of Formula (A-1-i): . In a second step, an ammoniacal treatment of the compound of Formula (A-1-i) is carried out, leading to the compound 005: . In a third step, the mono-phosphorylation of compound 005, in the presence of phosphoryl chloride and a trialkyl phosphate, leads to the phosphorodichloridate of Formula (B-i): In a fourth step, the phosphorodichloridate of Formula (B-i) is hydrolyzed to yield the compound 001: Alternatively, in a fifth step, the phosphate compound 001 obtained in the fourth step is then reacted, with the phosphorodichloridate compound of Formula (B-i) obtained as described in the third step, to give compound 009. According to one embodiment, a step of reducing compound 005 A-2 is carried out, leading to compound 007. The compound of formula 007 is then monophosphorylated as described in the fourth step and hydrolyzed to the compound 003. [0001] The above method for the preparation of the compounds 001, 003, 005 and 007 can be easily adapted to the synthesis of compounds 002, 004, 006 and 008 by using the suitable starting ribose tetraacetate of Formula (D-ii): The above method for the preparation of the dimer compound 009 can be easily adapted to the synthesis of dimer compounds 010-014 by using corresponding suitable phosphorodichloridate and phosphate intermediates. Pharmaceutical composition This invention also relates to a pharmaceutical composition for use in the stimulation of the expression of fetal hemoglobin comprising the compound for use according to the invention, and at least one pharmaceutically acceptable carrier. According to one embodiment, the pharmaceutical composition for use of the invention comprises, in addition to the at least one compound for use of the invention, at least one additional active ingredient, e.g., an active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics;  antioxidant such as CoQ10 and PQQ (Pyrroloquinoline quinone); hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab.  Non limiting examples of a natural extract are glycoproteins extract; terpenoids extract containing pentacyclic triterpenes such as betulin, pentacyclic triterpene metabolite such as betulinic acid, tramspiroins, rosenolactones, sesquiterpenes, erinacins; a flavonoid extract containing flavones, flavonols, flavanones, flavanols bioflavonoids or isolfavonoids; a polysaccharide extract containing PSP, PSK, CVG, HPB-3, H6PC20; or a polyaromatic molecule such as Hericerins and hericenones; from species such as Trametes versicolor, Hericium erinaceus, Grifola frondasa, milk thistle, artichoke, turmeric, dandelion, yellow dock, beetroot and ginger. More specifically, the composition of the invention increases the amount of fetal hemoglobin in the blood of a subject and/or the number of red blood cells expressing fetal hemoglobin and/or the proportions of red blood cells expressing fetal hemoglobin. The compositions of the invention are particularly useful for patients having sickle cell diseases, especially children. The compositions according to the invention may be formulated with carriers, excipients and diluents which are suitable in themselves for these formulations, such as lactose, dextrose, saccharose, sorbitol, mannitol, starches, acacia gum, calcium phosphate, alginates, tragacanth gum, gelatine, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, polyethylene glycol, cellulose, water (sterile), methyl cellulose, methyl and propyl hydroxybenzoates, talc, magnesium stearate, edible oils, vegetable and mineral oils or suitable mixtures thereof. The formulations may possibly contain other substances that are commonly used in pharmaceutical formulations, such as lubricants, wetting agents, emulsifying and suspending agents, dispersing agents, disintegrants, fillers, preservatives, sweeteners, flavourings, flow regulators, mould release agents, etc. The compositions can also be formulated to allow a rapid, prolonged or delayed release of the active compound(s) they contain. The compositions according to the invention are preferably in the form of unit doses and may be packaged in an appropriate manner, for example in a box, blister pack, bottle, sachet, ampoule or in any other suitable single-dose or multiple-dose carrier or container (which may be properly labelled); optionally with one or more leaflet(s) containing product information and/or instructions for use. Compositions of the invention can be prepared by any method known by the skilled in the art. Medical use and methods of treatment This invention thus relates to a compound of the invention as described hereinabove for use in stimulation of the expression of fetal hemoglobin. In other words, the compound of the invention as well as the composition of the invention comprising the compound increase the amount of fetal hemoglobin in the blood and/or in the red blood cells of a subject in need thereof and/or the proportion of RBC expressing fetal hemoglobin. Indeed, the inventors have observed that the administration of the compound of the invention to a patient increases the amount of fetal hemoglobin in the blood as well as the amount of fetal hemoglobin in the red blood cells of said patient. The level of fetal hemoglobin in the blood and in the red blood cells can be measured by any method known by the skilled in the art such as high performance ion-exchange liquid chromatography (HPLC) and immunofluorescence analysis by flow cytometry (N.Herbert et al, Individual red blood cell fetal hemoglobin quantification allows to determine protective thresholds in sickle cell disease, Am J Hematol.2020;95:1235–1245). The increase of the amount of fetal hemoglobin or the increase of the expression of fetal hemoglobin is a compared to baseline. The baseline can be the amount of fetal hemoglobin in RBC or the expression of fetal hemoglobin in RBC before administration of the compound of the invention. According to an embodiment, the compounds of the invention increase the amount of fetal hemoglobin in the blood of a subject and/or the number of RBCs expressing fetal hemoglobin. According to an embodiment, the amount of fetal globin in the blood of the subject increases. According to an embodiment, the percentage of fetal globin in the blood of the subject increases. According to an embodiment, the number of F-cells in the blood of the subject increases. According to an embodiment, the amount of total hemoglobin in the blood of the subject increases. According to an embodiment, the number of F- reticulocytes in the blood of the subject increases. By “F-cell”, it is referred to red blood cells expressing fetal-hemoglobin.   The stimulation of the expression of fetal hemoglobin can notably be used in the prevention or treatment of: - sickle cell disease and sickle cell disorders (sickle cell anemia with crisis, sickle cell anemia without crisis, double heterozygous sickling disorders, sickle cell trait and other sickle cell disorders), - thalassemia such as alpha-thalassemia, beta-thalassemia (for example, hemoglobin E beta thalassemia), delta-beta-thalassemia, thalassemia trait, hereditary persistence of fetal hemoglobin (HPFP), and other thalassemias, - coagulation defects (e.g., disseminated intravascular coagulation (difibrination syndrome)), hereditary factor VIII deficiency (hemophilia A), hereditary factor IX deficiency (Christmas disease), and other coagulation defects such as Von Willebrand's disease, hereditary factor Xi deficiency (hemophilia C), purpura (e.g., qualitative platelet defects and Glanzmann's disease), - neutropenia, agranulocytosis, functional disorders of polymorphonuclear neutrophils, other disorders of white blood cells (e.g., eosinophilia, leukocytosis, lymophocytosis, lymphopenia, monocytosis, and plasmacyclosis), - diseases of the spleen, methemoglobinemia, - other diseases of blood and blood forming organs (e.g., familial erythrocytosis, secondary polycythemia, essential thrombocytosis and basophilia), thrombocytopenia, hemoglobin C, D and E disease, hemoglobin lepore disease, and HbH and HbS diseases, anemias due to blood loss, radiation therapy or chemotherapy, or thrombocytopenias and neutropenias due to radiation therapy or chemotherapy, and certain diseases involving lymphoreticular tissue and reticulohistiocytic system (e.g., Langerhans cell hystiocytosis, eosinophilic granuloma, Hand-Schuller-Christian disease, hemophagocytic lymphohistiocytosis, and infection-associated hemophagocytic syndrome). Preferably, the subject has SCD or thalassemia, more preferably alpha-thalassemia or beta-thalassemia and even more preferably beta-thalassemia. By “sickle cell disease” (SCD) or “drepanocytosis” it is referred to a group of inherited red blood cell disorders defined by a missense point mutation in the sequence of beta globin, which results in a glutamic acid residue at position 6 being substituted by a valine. This mutated globin, called sickle hemoglobin or hemoglobin S (HbS), aggregates, and forms fibrous precipitates upon low oxygen level, leading to polymerized hemoglobin and promoting red blood cell (RBC) sickling. Non limiting examples of complications associated with sickle cell disease include acute chest syndrome, acute pain crisis, chronic pain, delayed growth and puberty, avascular necrosis, eye problems, gallstones, heart problems including coronary heart disease and pulmonary hypertension, infections such as meningitis, osteomyelitis, and sepsis; joint problems, kidney problems, leg ulcers, liver complications, pregnancy complications, priapism, severe anemia, stroke, renal necrosis or silent brain injury. The administration of the compound of the invention, by alleviating the symptoms and the severity of SCD, may prevent or treat the occurrence of complications SCD, or at the very least reduce the severity of the complications of SCD. By “beta-thalassemia”, it is referred to syndromes resulting from mutations, which cause a deficiency of beta globin chains. In beta thalassemia, the unmatched alpha globin chains aggregate inside red blood cells and their progenitors, causing the premature destruction of RBCs and RBC progenitors, which results in anemia, transfusion dependance, iron overload, organ failure and early death. It has been noted that patients expressing fetal hemoglobin experienced less complications than other patients with sickle cell disease or beta-thalassemia. Thus, according to one embodiment, the compound of the invention as described herein above is for use in the stimulation of the expression of fetal hemoglobin. The compound of the invention may be used in monotherapy or in combination therapy in a subject in need of therapeutic and/or preventive treatment. Thus, according to a first embodiment, the compound for use of the invention is administered to the subject without any other active ingredient. Thus, according to a second embodiment, the compound for use of the invention is administered to the subject in combination with at least one additional active ingredient, e.g., an active ingredient as described hereinabove. In one embodiment, the compound is administrated to the subject sequentially, simultaneously and/or separately with the other active ingredient as described hereinabove. The present invention also concerns a pharmaceutical composition comprising at least one compound for use of the invention, as described hereinabove, and at least one pharmaceutically acceptable carrier for use in the stimulation of the expression of fetal hemoglobin. Subjects in need of treatment Preferably, the subject in need of therapeutic and/or preventive treatment is a warm-blooded animal, more preferably a human. According to one embodiment, the subject is a male. According to one embodiment, the subject is a female. According to one embodiment, the subject is an adult, i.e. over 18 years of age. According to one embodiment, the subject is a child, i.e. under 18 years of age. According to one embodiment, the subject is an infant, i.e. having an age of more than one month and less than two years. According to one embodiment, the subject is a newborn, i.e. having an age from birth to less than one month. According to another preferred embodiment, the subject is of less than 20, 15, 10, 2 or 1 year(s) of age. In one embodiment, the subject is of less than 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 year(s) or 5 months of ages. According to one embodiment, said child is 10 or young than 10 years old, preferably 5 or young than 5 years old, more preferably 2 or younger than 2 years old. According to one embodiment, the subject does not suffer from any underlying pathology. According to one embodiment, the subject is at risk of developing sickle cell disease. According to one embodiment, the subject at risk of developing sickle cell disease belongs to an ethnic group selected from people of African descent, including African- Americans; Hispanic-Americans from Central and South America; People of Middle Eastern, southern European, Asian, Indian, and Mediterranean descent. According to one embodiment, the subject at risk of developing beta-thalassemia belongs to an ethnic group selected from people of Southeast Asian ancestry, such as Cambodians, Vietnamese, and Thai. According to one embodiment, the subject in need receives a treatment of at least one compound for use as described above at a cumulative dose of greater than 100 mg/kg, 200 mg/kg, 300 mg/kg, 400 mg/kg, 500 mg/kg, 600 mg/kg, 700 mg/kg, 800 mg/kg, 900 mg/kg or 1000 mg/kg. In one embodiment, the subject in need receives a treatment of at least one compound for use as described above at a cumulative dose of greater than 400 mg/kg, 500 mg/kg, 600 mg/kg, 700 mg/kg, 800 mg/kg, 900 mg/kg or 1000 mg/kg. According to one embodiment, the subject in need of therapeutic and/or preventive treatment is diagnosed by a health professional. For example, sickle cell disease may be diagnosed by various screening test routinely carried out in the medical setting, including newborn or prenatal screening, and aim to identify if the subject has abnormal hemoglobin genes in their red blood cells. This invention also relates to the use of a compound of invention or a pharmaceutical composition as described hereinabove in the stimulation of the expression of fetal hemoglobin as described hereinabove. This invention also relates to the use of a compound of the invention or a pharmaceutical composition as described hereinabove in the manufacture of a medicament for the stimulation of the expression of fetal hemoglobin as described hereinabove. This invention also relates to a method for the stimulation of the expression of fetal hemoglobin as described hereinabove in a subject in need thereof, comprising a step of administrating to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutical composition as described hereinabove. Another object of the invention is a kit-of-parts comprising a first part comprising a compound of the invention as described hereinabove, and a second part comprising another active ingredient, e.g., an active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ; hydroxyurea, L- glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. In one embodiment, the kit-of-parts of the invention comprises a first part comprising compounds 001-014, or a pharmaceutically acceptable salt or solvate thereof, and a second part comprising another active ingredient, e.g., an active ingredient as described hereinabove. Methods of administration The compounds of the invention as describes hereinabove, may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intracisternal injection or infusion, subcutaneous injection, or implant), by inhalation spray, nasal, rectal, sublingual, or topical routes of administration and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. In addition to the treatment of warm-blooded animals, such as mice, rats, horses, cattle, sheep, dogs, cats, monkeys, etc., the compounds of the invention are effective for use in humans. The pharmaceutical compositions for the administration of the compounds of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material, such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated by the techniques described in the U.S. Patents 4,256,108; 4,166,452; and 4,265,874 to form osmotic therapeutic tablets for control release. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil. In an embodiment, the composition is in the form of a gastro-resistant capsule or gastro- resistant tablet. Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol , such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin. Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant, such as ascorbic acid. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present. Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavouring and colouring agents. The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids, such as oleic acid find use in the preparation of injectables. The compounds of the present invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols. For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles.) Dosing regimen In the use for the stimulation of the expression of fetal hemoglobin, an appropriate dosage level will generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses. Preferably, the dosage level will be about 0.1 to about 350 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day. A suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg per day. For oral administration, the compositions are preferably provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The compounds may be administered on a regimen of 1 to 4 times per day, preferably once, twice or three times per day. It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy. Monotherapy/Combination therapy The nicotinamide mononucleotide derivatives of the invention may be used in monotherapy or in combination therapy in a subject in need of therapeutic and/or preventive treatment. Thus, according to a first embodiment, the compound for use of the invention is administered to the subject without any other active ingredient. According to a second embodiment, the compound for use of the invention is administered to the subject in combination with at least one additional active ingredient, e.g., an active ingredient as described hereinabove. In one embodiment, the compound is administrated to the subject sequentially, simultaneously and/or separately with the other active ingredient. In one embodiment, the other active ingredient is selected from natural extracts; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ; hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. According to one embodiment, the pharmaceutical composition of the invention further comprises at least another active ingredient. According to one embodiment, the pharmaceutical composition for use of the invention comprises, in addition to the at least one compound for use of the invention, at least one additional active ingredient, e.g., an active ingredient selected from natural extracts; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ; hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. According to one embodiment, the compound of the invention is used in combination with blood transfusion, especially red blood cell transfusion. In one embodiment, the compound of the invention is administrated to the subject sequentially, simultaneously and/or separately with the blood transfusion. Kit of parts Another object of the invention is a kit-of-parts comprising a first part comprising a compound of the invention as described hereinabove, and a second part comprising another active ingredient, e.g., an active ingredient selected from but not limited to a natural extract; opioid or non-opioid analgesics; NSAIDS; antidepressants; anticonvulsants; antibiotics; antioxidant such as CoQ10 and PQQ; hydroxyurea, L-glutamine, Kynurenine, kynurenic acid, tryptophan, Voxelator and Crizanlizumab. In one embodiment, the kit-of-parts of the invention comprises a first part comprising compounds 001-014, or a pharmaceutically acceptable salt or solvate thereof, and a second part comprising another active ingredient, e.g., an active ingredient as described hereinabove. Method of treatment This invention also relates to the use of a compound of invention or a pharmaceutical composition as described hereinabove in the treatment of a red blood cell disorder as described hereinabove. This invention also relates to the use of a compound of the invention or a pharmaceutical composition as described hereinabove in the manufacture of a medicament for the treatment of a red blood cell disorder as described hereinabove. This invention also relates to a method for the treatment of a red blood cell disorder as described hereinabove in a subject in need thereof, comprising a step of administrating to said subject a therapeutically effective amount of a compound of the invention or a pharmaceutical composition as described hereinabove. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 is a histogram showing the percentage of RBC expressing fetal hemoglobin (F- cells) using antibodies against fetal hemoglobin by flow cytometry (FACS). Figure 2 is a histogram showing fetal hemoglobin mean fluorescence intensity (MFI) evaluated by flow cytometry. Figure 3 is a histogram showing the reticulocyte counts using Reticount by flow cytometry. Figure 4 is a histogram showing the ability of compound 001 to prevent sickling of SS RBCs at a 1% O 2 . Figure 5 is a histogram showing the ability of compounds 010 to prevent sickling of SS RBCs at a 1% O 2 . Figure 6 is a histogram showing the ability of compounds 011 to prevent sickling of SS RBCs at a 1% O 2 . Figure 7 is a histogram showing the percentage of F-cells using antibodies against fetal hemoglobin by flow cytometry after treatment by compound 001 and/or L-glutamine. Figure 8 is a histogram showing the reticulocyte counts of red blood cells expressing fetal hemoglobin using Reticount by flow cytometry, after treatment by compound 001 and/or L-glutamine. Figure 9 is a dot plot showing the percentage of RBC expressing fetal hemoglobin (F- cells). Figure 10 is a dot plot showing the fetal hemoglobin mean fluorescence intensity (MFI) evaluated by flow cytometry. EXAMPLES The present invention is further illustrated by the following examples, which are provided for the purpose of illustrating the invention without limiting the invention to the sole embodiments disclosed below. Example 1: Synthesis of compounds of the invention Materials and Methods All materials were obtained from commercial suppliers and used without further purification. Thin-layer chromatography was performed on TLC plastic sheets of silica gel 60F254 (layer thickness 0.2 mm) from Merck. Column chromatography purification was carried out on silica gel 60 (70-230 mesh ASTM, Merck). Melting points were determined either on a digital melting point apparatus (Electrothermal IA 8103) and are uncorrected or on a Kofler bench type WME (Wagner & Munz). IR, 1 H, 19 F and 13 C NMR spectra confirmed the structures of all compounds. IR spectra were recorded on a Perkin Elmer Spectrum 100 FT-IR spectrometer and NMR spectra were recorded, using CDCl 3 , CD 3 CN, D 2 O or DMSO-d 6 as solvent, on a Bruker AC 300, Advance DRX 400 and Advance DRX 500 spectrometers, for 1 H, 75 or 100 MHz for 13 C and 282 or 377 MHz for 19 F spectra. Chemical shifts (δ) were expressed in parts per million relative to the signal indirectly (i) to CHCl 3 (δ 7.27) for 1 H and (ii) to CDCl 3 (δ 77.2) for 13 C and directly (iii) to CFCl 3 (internal standard) (δ 0) for 19 F. Chemical shifts are given in ppm and peak multiplicities are designated as follows: s, singlet; br s, broad singlet; d, doublet; dd, doublet of doublet; t, triplet; q, quadruplet; quint, quintuplet; m, multiplet. The high-resolution mass spectra (HRMS) were obtained from the "Service central d'analyse de Solaize" (Centre national de la recherche scientifique) and were recorded on a Waters spectrometer using electrospray-TOF ionization (ESI-TOF). General experimental procedures Step 1: Synthesis of the compound of formula A-1 The compound of formula D (1.0 equiv.) is dissolved in dichloromethane. Nicotinamide of formula E (1.50 equiv.) and TMSOTf (1.55 equiv.) are added at room temperature. The reaction mixture is heated under reflux and stirred until the reaction is complete. The mixture is cooled to room temperature and filtered. The filtrate is concentrated to dryness to give tetraacetate A-1. Step 2: Synthesis of the compound of formula A-2 Tetraacetate A-1 is dissolved in methanol and cooled to -10 °C. Ammonia 4,6 M in methanol (3,0 equivalents) at -10 °C is added and the mixture is stirred at this temperature until the reaction is complete. Dowex HCR (H+) resin is added up to pH 6-7. The reaction mixture is heated to 0 °C and filtered. The resin is washed with a mixture of methanol and acetonitrile. The filtrate is concentrated to dryness. The residue is dissolved in the acetonitrile and concentrated to dryness. The residue is dissolved in the acetonitrile to give a solution of the compound of formula A-2. Step 3: Synthesis of the compound of formula A-3 The solution of the crude compound of formula A-2 in acetonitrile is diluted with trimethyl phosphate (10.0 equivalents). The acetonitrile is distilled under vacuum and the mixture is cooled to -10 °C. Phosphorus oxychloride (4,0 equivalents) is added at 10 °C and the mixture is stirred at 10 °C until the reaction is complete. Steps 4 and 5: Synthesis of the compound of formula 001 The mixture obtained in step 3 above is hydrolyzed by the addition of a 50/50 mixture of acetonitrile and water, followed by the addition of methyl tert-butyl ether. The mixture is filtered and the solid is dissolved in water. The aqueous solution is neutralized by the addition of sodium bicarbonate and extracted with dichloromethane. The aqueous layer is concentrated to dryness to yield the crude formula 001 compound, which is purified on a DOWEX 50wx8 column with elution in water followed by a silica gel chromatographic column. Step 4 and step 5: Synthesis of compound of formula 009 The mixture is hydrolyzed by addition of a 50/50 mixture of acetonitrile and water, followed by addition of tert-butyl methyl ether. The mixture is filtered and the solid is dissolved in water. The aqueous solution is neutralized by addition of sodium bicarbonate and extracted with dichloromethane. The aqueous layer is concentrated to dryness to give a crude mixture of NMN and di-NMN of formula 009. Isolation of di-NMN of formula 009: NMN and di-NMN of formula 009 are separated by purification on Dowex 50wx8 with water elution. The fractions containing di-NMN are concentrated to dryness. The residue is purified by column chromatography on silica gel (gradient isopropanol/water). Pure fractions are combined and concentrated. The residue is freeze-dried to afford di-NMN as a beige solid. 31 P RMN : δ (ppm, reference 85% H 3 PO 4 : 0 ppm dans D 2 O) = -11.72 ; 1 H RMN : δ (ppm, reference TMS: 0 ppm dans D 2 O) = 4.20 (ddd, J H-H = 11.9, 3.5, 2.4 Hz, 2H), 4,35 (ddd, J H-H = 11.9, 3.9, 2.2 Hz, 2H), 4.43 (dd, J H-H = 5,0, 2.6 Hz, 2H), 4.53 (t, J H-H = 5.0 Hz, 2H), 4.59 (m, 2H), 6.16 (d, J H-H = 5.4 Hz, 2H), 8.26 (dd, J H-H = 8.1, 6.3 Hz, 2H), 8.93 (d, J H-H = 8.1 Hz, 2H), 9.25 (d, J H-H = 6.2 Hz, 2H), 9.41 (s, 2H) ; 13 C RMN : δ (ppm, reference TMS: 0 ppm dans D 2 O) = 64.84 (CH 2 ), 70.73 (CH), 77.52 (CH), 87.11 (CH), 99.88 (CH), 128.65 (CH), 133.89 (Cq), 139.84 (CH), 142.54 (CH), 146.04 (CH), 165.64 (Cq); MS (ES+) : m/z = 122.8 [Mnicotinamide + H]+, 650.8 [M + H]+. Synthesis of compound of formula 010 Phosphorus oxychloride (3.0 eq.) is added to trimethylphosphate (20.0 eq.) at -5°C. β-NR chloride (1.0 eq.) is added by portions at -5°C and the reaction mixture stirred overnight at -5°C. Morpholine (3.0 eq.) is added dropwise at -10/0°C and the mixture stirred for 2- 3 h. a-NMN (1.0 eq.) is then added by portions at -5°C and the reaction mixture stirred at -5°C overnight. Hydrolysis is performed by dropwise addition of water (5 vol.) at -10/0°C and the mixture is stirred until complete homogenization at 10-15°C. The reaction mixture is then extracted with dichloromethane (6*10 vol.) and the aqueous phase neutralized by eluting through Purolite A600E formate form resin (theoretical amount to neutralize HCl coming from POCl 3 ). The eluate is then concentrated on vacuum at 45/50°C to give the crude containing the α,β-diNMN of formula 010. Elution with water through Dowex 50wx8100-200 mesh H + form resin allows removing of some impurities. Fractions containing compound 010 are combined and concentrated on vacuum at 45- 50°C. The crude is then purified by preparative chromatography on Luna Polar RP 10µm stationary phase with elution with a 10mM NaH 2 PO 4 aqueous solution. Pure fractions are combined and eluted with water on Purolite C100EH H + form resin (needed quantity to fully exchange Na + by H + ), then eluted on Purolite A600E acetate form resin (needed quantity to fully exchange H2PO4- by acetate). The eluate is concentrated on vacuum and the residue freeze-dried to afford compound 010 as a white solid. 31 P RMN : δ (ppm, reference 85% H 3 PO 4 : 0 ppm dans D 2 O) = -11.87, -11.69, -11.46, - 11.29; 1 H RMN : δ (ppm, reference TMS: 0 ppm dans D 2 O) = 4.10 (ddd, J = 11.1, 6.1, 3.1 Hz,1H), 4.15-4.25 (m, 2H), 4.36 (ddd, J = 12.2, 4.4, 2.4 Hz, 1H), 4.40 (dd, J = 4.9, 2.4 Hz, 1H), 4.44 (dd, J = 5.0, 2.7 Hz, 1H), 4.53 (t, J = 5.0 Hz, 1H), 4.5 (m, 1H), 4.85 (m, 1H), 4.92 (t, J = 5.3 Hz, 1H), 6.15 (d, J = 5.5 Hz, 1H), 6.51 (d, J = 5.7 Hz, 1H), 8.14 (dd, J = 8.0, 6.3 Hz, 1H), 8.26 (dd, J = 8.1, 6.3 Hz, 1H), 8.88 (d, J = 8.1 Hz, 1H), 8.92 (d, J = 8.1 Hz, 1H), 9.02 (d, J = 6.3 Hz, 1H), 9.24 (s, 1H), 9.26 (d, J = 6.4 Hz, 1H), 9.40 (s, 1H); 13 C RMN : δ (ppm, reference TMS: 0 ppm dans D 2 O) = 64.83, 64.87 (CH 2 ), 65.30, 65.35 (CH 2 ), 70.65 (CH), 70.74 (CH), 71.92 (CH), 77.51 (CH), 87.03, 87.10 (CH), 87.19, 87.26 (CH), 96.57 (CH), 99.83 (CH), 126.89 (CH), 128.54 (CH), 132.44 (Cq), 133.81 (Cq), 139.85 (CH), 140.92 (CH), 142.50 (CH), 143.49 (CH), 145.06 (CH), 145.97 (CH), 165.64 (Cq), 165.88 (Cq); MS (ES+) : m/z = 122.8 [Mnicotinamide + H]+, 650.9 [M + H]+. Synthesis of compound of formula 011 Phosphorus oxychloride (3.0 eq.) is added to trimethylphosphate (20.0 eq.) at -5°C. α-NR chloride (1.0 eq.) is added by portions at -5°C and the reaction mixture stirred overnight at -5°C. Morpholine (3.0 eq.) is added dropwise at -10/0°C and the mixture stirred for 2- 3 h. α-NMN (1.0 eq.) is then added by portions at -5°C and the reaction mixture stirred at -5°C overnight. Hydrolysis is performed by dropwise addition of water (5 vol.) at -10/0°C and the mixture is stirred until complete homogenization at 10-15°C. The reaction mixture is then extracted with dichloromethane (6*10 vol.) and the aqueous phase neutralized by eluting through Purolite A600E formate form resin (theoretical amount to neutralize HCl coming from POCl 3 ). The eluate is then concentrated on vacuum at 45/50°C to give the crude containing the α,α-diNMN of formula 011. Elution with water through Dowex 50wx8100-200 mesh H + form resin allows removing of some impurities. Fractions containing the compound 011 are combined and concentrated on vacuum at 45- 50°C. The crude is then purified by preparative chromatography on Luna Polar RP 10µm stationary phase with elution with a 10mM NaH2PO4 aqueous solution. Pure fractions are combined and eluted with water on Purolite C100EH H + form resin (needed quantity to fully exchange Na + by H + ), then eluted on Purolite A600E acetate form resin (needed quantity to fully exchange H 2 PO 4 - by acetate). The eluate is concentrated on vacuum and the residue freeze-dried to afford compound 011 as a white solid. 31 P RMN : δ (ppm, reference 85% H 3 PO 4 : 0 ppm dans D 2 O) = -11.40; 1 H RMN : δ (ppm, reference TMS: 0 ppm dans D 2 O) = 4.14 (ddd, J = 11.4, 3.4, 2.8 Hz, 2H), 4.23 (ddd, J = 11.6, 3.3, 2.8 Hz, 2H), 4.44 (dd, J = 4.8, 2.3 Hz, 2H), 4.88 (m, 2H), 4.96 (t, J = 5.3 Hz, 2H), 6.54 (d , J = 5.7 Hz, 2H), 8.15 (dd, J = 8.1, 6.2 Hz, 2H), 8.89 (d, J = 8.1 Hz, 2H), 9.05 (d, J = 6.3 Hz, 2H), 9.26 (s, 2H); 13 C RMN : δ (ppm, reference TMS: 0 ppm dans D2O) = 65.37 (CH 2 ), 70.70 (CH), 71.95 (CH), 87.30 (CH), 96.62 (CH), 126.91 (CH), 132.45 (Cq), 140.94 (CH), 143.52 (CH), 145.07 (CH),165.90 (Cq); MS (ES+) : m/z = 122.7[Mnicotinamide + H]+, 650.8 [M + H]+. Example 2: Evaluation of compounds of the invention on sickle red blood cell experimental models The aim of the present study was to evaluate, the effects of i.p. daily administration of compounds 001, 010 and 011 at 185 mg/kg as modulator of red blood cell sickling and fetal hemoglobin expression in erythroid cells and its potential role in therapy for sickle cell disease on mouse model of SCD. I. Materials and Methods Material Animals: Townes S/S mice on a 129/B6 mixed genetic background. Methods 1. Preparation of formulation: The powder of compounds 001, 010 and 011 (185 mg/kg) were dissolved in vehicle (the solution is used at room temperature for maximum 1 day). A fresh sample for each administration was prepared every day except the week-end (the solution is prepared on Saturday and is used on Saturday and Sunday). 2. Sickle red blood cell In Townes S/S mice, mouse alpha-and beta- globin gene loci are deleted and replaced by human alpha and beta globins. When carrying two copies of the beta S allele, mice develop a human sickle disease phenotype with sickle-shaped red blood cells are seen in blood smears. 3. Experimental groups Group description : Group 1: Vehicle (i.p.) Group 2: compound 001 (185 mg/kg) Group 3 compound 010 (185 mg/kg) Group 4: compound 011 (185 mg/kg) 4. Treatment Mice were i.p treated with compounds 001, 010 and 011 during all the experiment (D0 to D15) once per day. Last injection occurred 24 hours before sacrifice. 5. Blood collection Retro-orbital blood collection was performed at the inclusion D0 and at D5, D10 and D15. 6. Ex-vivo Ex-vivo blood collected were assessed for percentage of F-cells using antibodies against fetal hemoglobin and reticulocyte counts using reticount by flow cytometry (FACS). Red blood cells sickling was assessed under hypoxia. II. Results and discussion 1. Percentage of F-Cells The figure 1 shows the percentage of F-cells using antibodies against fetal hemoglobin by flow cytometry (FACS) . The results show that treatment with: - Compound 001 (185 mg/kg/d, i.p.) led to a significant increase of the mean F-cells from less than 5% to 8% over the 15 days treatment of mice (figure 1); Figure 2 shows fetal hemoglobin mean fluorescence intensity (MFI) evaluated by flow cytometry: - A non-significant increase was observed on fetal hemoglobin median fluorescence intensity (MFI) per cell at day 15 (300.4 ±13.3 MFI at day 0; 290.6 ±10.7 MFI p=0.9690 at day 5; 295.4 ±19.4 MFI p=0.9956 at day 10; 345.4 ±22.9 MFI p=0.2089 at day 15). 2. Reticulocyte counts using reticount The figure 3 shows the reticulocyte counts using Reticount by flow cytometry (FACS). The results show that treatment with: - Compound 001 (185 mg/kg/d, i.p.) led to a significant decrease of the percentage of reticulocytes from 70% to 30% over the 15 days treatment of mice (figure 2); 3. RBC sickling under hypoxia ex vivo Figures 4, 5 and 6 show the ability of compounds 001 (Fig.3), 010 (Fig.4) and 011 (Fig. 5) to prevent sickling of SS RBCs at a 1% O 2 . SS RBCs from treated mice and collected at D0, D5, D10 and D15 were submitted to hypoxia for 30 minutes in a hypoxic chamber (1% O 2 ). Percentage of sickling RBCs was then assessed for each time point with compounds 001, 010 and 011. The results showed that treatment with: - Compound 001 (185 mg/kg/d, i.p.) led to a decrease of the percentage of sickling cells from 40% at D0 to less than 10% after 15 days treatment of mice (figure 4). - Compound 010 (185 mg/kg/d, i.p.) led to a significant decrease of the percentage of sickling cells from 32% at D0 to less than 15% after 15 days treatment of mice. - Compound 011 (185 mg/kg/d, i.p.) led to a significant decrease of the percentage of sickling cells from 31% at D0 to 20% after 15 days treatment of mice. III. Conclusion These results indicate that treatments with compounds 001, 010 and/or 011 reduce red blood cell sickling under hypoxia and increase fetal hemoglobin expression in erythroid cells illustrating their potential role in therapy for sickle cell disease. Example 3 – Use of compound 001 (beta-NMN) and/or L-Glutamine to increase fetal hemoglobin The purpose of this study is to evaluate the effects of i.p. daily administration of compounds 001 at 185 mg/kg and/or L-Glutamine (L-Gln) on the level of fetal hemoglobin expression. L-Gln is standard of care for sickle cell disease (SCD) patients in the United States as it has been shown that L-Gln administration reduces the severity and frequency of VOCs. IV. Materials and Methods Material Animals: Townes S/S mice on a 129/B6 mixed genetic background aged 8-12 weeks. Methods 1. Preparation of formulation: The powder of compound 001 (185 mg/kg) was dissolved in vehicle (the solution is used at room temperature for maximum 1 day). The powder of L-Glutamine (180 mg/kg) was dissolved in vehicle (the solution is used at room temperature for maximum 1 day). A fresh sample for each administration was prepared every day except the weekend (the solution is prepared on Saturday and is used on Saturday and Sunday). 2. Sickle red blood cell In Townes S/S mice, mouse alpha- and beta-globin gene loci are deleted and replaced by human genes coding for the alpha- and beta- globins. When carrying two copies of the beta S allele, mice develop a human sickle disease phenotype with sickle-shaped red blood cells are seen in blood smears. 3. Experimental groups Group description : Group 1: Vehicle PBS (i.p.) Group 2: Compound 001 (185 mg/kg) Group 3 L-Gln (180 mg/kg) Group 4: Compound 001 (185 mg/kg) + L-Gln (180 mg/kg) 4. Treatment Mice were i.p treated with Compound 001, L-Gln or the combination of Compound 001+L-Gln during all the experiment (D0 to D15) once per day. Last injection occurred 24 hours before sacrifice. 5. Blood collection Retro-orbital blood collection was performed at the inclusion D0 and at D5, D10 and D15. 6. Ex-vivo Ex-vivo blood collected were assessed for percentage of F-cells using antibodies against fetal hemoglobin and reticulocyte counts using reticount by flow cytometry. The amount of fetal hemoglobin has been assessed under normoxia and hypoxia. V. Results and discussion 4. Percentage of F-Cells Figure 7 shows the percentage of F-cells using antibodies against fetal hemoglobin by flow cytometry. The results show that treatment with: - As expected, L-Gln increases the percentage of F-cell carrying fetal hemoglobin compared to Vehicle in hypoxia. - Compound 001 (185 mg/kg/d, i.p.) led to a significant increase of the mean F-cells percentage in both normoxia and hypoxia, compared to Vehicle. It also provided a significant increase compared to the groups treated by Vehicle or L-Gln. - The combination of Compound 001 and L-Gln does not improve the results obtained with compound 001 alone. 5. Fetal hemoglobin content in F-cells Figure 8 shows the number of mean fluorescence intensity measured in F-cells by flow cytometry. The results show that treatment with: - As expected, L-Gln increases the amount of fetal hemoglobin in RBC compared to Vehicle in hypoxia. - Compound 001 (185 mg/kg/d, i.p.) led to a significant increase of the content of HbF in F-cells in both normoxia and hypoxia, compared to Vehicle, as shown by increased MFI. It also provided a significant increase compared to the groups treated by Vehicle or L-Gln. - The combination of Compound 001 and L-Glutamine does not improve the results obtained with compound 001 alone. Therefore, the inventors have demonstrated that the compound of formula I according to the invention can increase the amount of fetal hemoglobin in the blood of a subject, especially a subject having sickle cell disease, in both normoxic and hypoxic conditions. The compound of the invention is thus at least as efficient as L-Gln, the standard of care in the USA for sickle cell disease. Example 4 – Evaluation of Compound 001 (beta-NMN) to increase fetal hemoglobin over 28 days. Animals were dosed once daily, intra-peritoneally, for 28 days at a fixed dose volume of 200 µl per mouse, which was based on a nominal 20g body weight. Formulation of the test item Compound 001 (beta-NMN) was dissolved in PBS. The formulation was divided into 1 ml aliquots which were stored at -80°C and protected from light. For each day of dosing, an aliquot of each test item was removed from frozen storage. Residual thawed solutions were discarded on completion of each day of dosing. Animal model The animals used in this study Townes HbSS have been previously described (Wu LC et al. Blood 2006;108:1183-1188) and this genotype mimics the genetic, hematologic and histopathologic features that are found in humans afflicted with sickle cell disease. The stock background of this strain is a C57BL/6j-129/Sv genome. At the start of dosing, mice were in the weight range of 19.0-21.0 g and, on completion of dosing, mice weighed 19.0-22.5 g. Environment All mice were conventionally housed up to 5 per cage in solid bottomed plastic cages with corncob bedding. The animal room was maintained at temperature of 22–24 °C and a relative humidity of between 45 and 65%, with a 12-hour light/dark cycle. A pelleted laboratory diet and water (in bottles) were freely available. The Animal Care Committee at Paris Descartes University approved this mouse experiment. Fetal hemoglobin immunofluorescence analysis by flow cytometry. Blood obtained from mice treated for 0, 5, 10, 15 or 28 were washed in cold PBS and centrifuged for 5 minutes at 300 g. Cells were stained with antibody against TER-119 (Biolegend clone Ter-119 Pacific Blue #116232) for 30 minutes at 4°C washed with FACS staining buffer. Cells were fixed in 0.05% glutaraldehyde at room temperature for 10 minutes, and then in 0.1% Triton X-100 at room temperature for 10 minutes to allow membrane permeabilization. Cells were further washed with BD Perm/Wash™ buffer (BD Biosciences reference 554723). Cell suspensions were then stained with antibodies against fetal hemoglobin (PE-mouse anti-human fetal hemoglobin, clone 2D12, dilution 1/25, BD Biosciences reference 560041) or the isotype control (PE-mouse IgG1κ, clone MOPC-21, dilution 1/25, BD Biosciences reference 554680) for 30 minutes on ice and protected from light. Cells were further analyzed by flow cytometry (Gallios TM , Beckman) using FlowJo software v.10 (FlowJo, LLC). Statistical analysis Data were processed to give group mean values and standard deviations. Statistical analyses were performed using a one-way analysis of variance (ANOVA, ex vivo dataset) or Mann-Whitney test (in vivo dataset) test for analysis of treatment effect vs. controls. All statistical analyses and graphical output of results with applied statistics were derived using GraphPad software (v9.00, San Diego California USA). Probability values of less than 5% were regarded as providing sufficient evidence to reject the null hypothesis and therefore statistical significance was identified at the p<0.05 levels. Results Figure 9 shows the percentage of red blood cells expressing fetal hemoglobin. F-cells (percentage of cells expressing fetal hemoglobin) were increased on mice treated with Compound 001 for 28 days (3.5 ±0.6%, p<0.05) when compared to F-cell frequency of mice given vehicle (1.4 ±0.3%). Figure 10 shows fetal hemoglobin mean fluorescence intensity (MFI) evaluated by flow cytometry. Changes were observed on fetal hemoglobin median fluorescence intensity (MFI) per cell (increase of 29%): 1425 ±45 MFI at Compound 001-treated group vs. 1104 ±93 MFI on control group, p<0.01. Conclusion Over 28 days treatment, Compound 001 significantly increased the expression of fetal hemoglobin in red blood cells as shown by the increased mean fluorescence intensity in individual red blood cells and the increased proportion of red blood cells carrying fetal hemoglobin. * The compounds of the invention can thus be used to stimulate the expression of fetal hemoglobin in red blood cells in a patient.