WO2022162409A1 - Methods of treatment with s1p receptor modulators - Google Patents

Methods of treatment with s1p receptor modulators Download PDF

Info

Publication number
WO2022162409A1
WO2022162409A1 PCT/IB2021/000033 IB2021000033W WO2022162409A1 WO 2022162409 A1 WO2022162409 A1 WO 2022162409A1 IB 2021000033 W IB2021000033 W IB 2021000033W WO 2022162409 A1 WO2022162409 A1 WO 2022162409A1
Authority
WO
WIPO (PCT)
Prior art keywords
receptor modulator
mocravimod
hsct
unit
day
Prior art date
Application number
PCT/IB2021/000033
Other languages
English (en)
French (fr)
Inventor
Christoph Bucher
Simone DERTSCHNIG
Original Assignee
Priothera Sas
Priothera Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Priothera Sas, Priothera Limited filed Critical Priothera Sas
Priority to AU2021424131A priority Critical patent/AU2021424131A1/en
Priority to PCT/IB2021/000033 priority patent/WO2022162409A1/en
Priority to PCT/EP2022/051940 priority patent/WO2022162088A2/en
Priority to TW111103784A priority patent/TW202245741A/zh
Priority to KR1020237025402A priority patent/KR20230136603A/ko
Priority to CA3208122A priority patent/CA3208122A1/en
Priority to EP23191736.0A priority patent/EP4248958A3/en
Priority to MX2023008907A priority patent/MX2023008907A/es
Priority to AU2022214268A priority patent/AU2022214268A1/en
Priority to JP2023545210A priority patent/JP2024506825A/ja
Priority to EP22702938.6A priority patent/EP4284348A2/en
Priority to CN202280012135.2A priority patent/CN116723835A/zh
Publication of WO2022162409A1 publication Critical patent/WO2022162409A1/en
Priority to US18/066,883 priority patent/US20230130403A1/en
Priority to IL304729A priority patent/IL304729A/en
Priority to CL2023002212A priority patent/CL2023002212A1/es
Priority to CONC2023/0009893A priority patent/CO2023009893A2/es

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/397Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having four-membered rings, e.g. azetidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • A61K38/13Cyclosporins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection

Definitions

  • the present invention relates to S1 P receptor modulators, preferably mocravimod, for use in treating patients suffering from acute myelogenous leukemia and who have undergone allogeneic hematopoietic stem cell transplantation.
  • AML Acute myeloid leukemia
  • AML Acute myeloid leukemia
  • AML Acute myeloid leukemia
  • AML is an aggressive, fast-growing, disease in which too many myeloblasts (immature white blood cells that are not lymphoblasts) are found in the bone marrow and blood.
  • myeloblasts Immature white blood cells that are not lymphoblasts
  • AML acute myeloblastic leukemia
  • AML acute myelogenous leukemia
  • AML acute nonlymphocytic leukemia
  • ANLL ANLL
  • AML results when bone marrow begins making blasts instead of mature white blood cells. The immature blasts are unable to fight infections.
  • AML is the most common acute leukemia, and it progresses rapidly. Left untreated, AML may lead to death within weeks or months.
  • AML Advanced leukemia
  • chemotherapy including targeted chemotherapies, radiation therapy, and autologous hematopoietic stem cell transplantation or allogeneic hematopoietic stem cell transplantation.
  • the side effects of each of these treatments are well documented.
  • a newly diagnosed AML patient will be treated with an induction chemotherapy regimen to attempt to put the cancer in remission.
  • Remission is most often a temporary measure as most AML patients in remission eventually relapse.
  • Post-remission therapies include consolidation chemotherapy, allogenic hematopoietic stem cell transplantation, or autologous hematopoietic stem cell transplantation.
  • HSCT hematopoietic stem cell transplantation
  • major complications of HSCT include graft-versus-host disease (GVHD) and other life-threatening complications.
  • GVHD graft-versus-host disease
  • immunosuppressive prophylaxis 30 to 60% of patients will develop some level of acute GVHD (Abo-Zena and Horwitz, Curr Opin Pharmacol. 2002 Aug;2(4):452-7; Jagasia et al., Blood 2012 Jan 5;119(1 ):296-307; Ruggeri et al., J Hematol Oncol. 2016 Sep 17;9(1 ):89).
  • Immunosuppressive GVHD prophylaxis is based for example on Ciclosporin A (CsA) which is a Calcineurin inhibitor (CNI) and Methotrexate (MTX) as a well established immunosuppressive regimen.
  • CsA Ciclosporin A
  • CNI Calcineurin inhibitor
  • MTX Methotrexate
  • Criteria for decision making to proceed to allo-HSCT in patients in first Complete Remission (CR) include risk group classification at diagnosis based on prognostic factors, and donor availability, at the time of induction. Some criteria are added at the time of consolidation, such as the quality of remission and persistence of Measurable Residual Disease (MRD), co-morbidities (HCT-CI as index of comorbidity) and recipient-donor profile (Gratmple score).
  • EPN European Leukemia Network
  • the ELN has issued guidance for allo-HSCT eligibility in patients with intermed iate/adverse risk and an additional stratification to assess post remission therapies: Low, Intermediate, High risk of relapse (Dohner et al., Blood 2017 Jan 26;129(4):424-447).
  • HSCT is indicated for patients in first CR with adverse cytogenetic/ molecular characteristics and/ or high MRD, provided HCT-CI is good enough and provided a donor is available in a timely manner.
  • Chronic GVHD While acute GVHD is an inflammatory disease, chronic GVHD resembles an autoimmune syndrome and typically develops 4 to 6 months posttransplant due to antigen-specific donor immune cells that cause autoimmune clinical manifestations.
  • Chronic GVHD can occur independently from acute GVHD as the classic form or as an overlap syndrome with the presence of acute features.
  • the pathology of acute GVHD is characterized by T-cell infiltration causing tissue inflammation and damage and is a risk factor for the later development of chronic GVHD, which is a fibrotic and sclerotic disease. It remains, however, unknown if acute and chronic GVHD are directly connected, and if so, how they are mechanistically linked.
  • Sphingosine-1 -phosphate is a metabolite of sphingolipid, a component of biomembrane.
  • S1 P acts as a ligand for a family of five G-protein-coupled receptors, named S1 P receptor types 1 to 5 (S1 PR1 -5).
  • FTY720 a multi-S1 PR inhibitor of S1 PR1 , 3-5
  • HSCT allogeneic hematopoietic stem cell transplantation
  • Calcineurin inhibitors such as CsA and tacrolimus (TAC) suppress donor T-cell activation and remain the most commonly used immunosuppressants for acute GVHD prophylaxis.
  • W02020/022507 discloses the results of a clinical study for treating patients with hematological malignancies, where said patients undergo HSCT and are treated with KRP203 daily for a maximum duration of 110 days, together with standard of care to prevent acute GVHD with immunosuppressants, such as CsA and MTX, or MTX and TAC.
  • the invention described here meets this need, as the described methods provide a synergistic therapeutic effect, preventing acute and chronic GVHD effects while further improving GVL effect and risk of relapse in high-risk AML patients.
  • a first object of the invention relates to an S1 P receptor modulator, preferably mocravimod, for use in treating a human subject suffering from acute myelogenous leukemia (AML) and undergoing allogeneic hematopoietic stem cell transplant (HSCT).
  • AML acute myelogenous leukemia
  • HSCT allogeneic hematopoietic stem cell transplant
  • Another object of the invention relates to an S1 P receptor modulator, preferably mocravimod, for use in treating chronic GVHD, typically for delaying chronic GVHD onset, in a human subject undergoing allogeneic hematopoietic stem cell transplant (HSCT), preferably in a human subject in need of HSCT for treating acute myelogenous leukemia.
  • S1 P receptor modulator preferably mocravimod
  • Another object of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising an S1 P receptor modulator of formula (II): or pharmaceutically acceptable salts thereof, and at least one filler selected from mannitol, microcrystalline cellulose and mixtures thereof, sodium starch glycolate as disintegrant, magnesium stearate as lubricant and, colloidal silicon dioxide as glidant.
  • the present disclosure relates in particular to various methods of treating acute myelogenous leukemia in a subject undergoing allogeneic hematopoietic stem cell transplant (HSCT) comprising:
  • conditioning said subject for destroying substantially the bone marrow and immune system wherein said conditioning includes treatment of said subject with an effective amount of a chemotherapeutic agent and/or performing total body irradiation;
  • the present disclosure further relates to a method of preventing chronic GvHD in a subject undergoing allogeneic hematopoietic stem cell transplant (HSCT) comprising:
  • conditioning said subject for destroying substantially the bone marrow and immune system wherein said conditioning includes treatment of said subject with an effective amount of a chemotherapeutic agent and/or performing total body irradiation;
  • the disclosure further relates to the use of an S1 P receptor modulator in the manufacture of a medicament for treating acute myelogenous leukemia in a subject undergoing allogenic hematopoietic stem cell transplant.
  • the disclosure also relates to the use of an S1 P receptor modulator in the manufacture of a medicament for preventing chronic GVHD, typically delaying chronic GVHD onset, in a patient undergoing allogeneic hematopoietic stem cell transplant.
  • the S1 P receptor modulator is selected among mocravimod, FTY720, siponimod, fingolimod, ozanimod, ponesimod, etrasimod, AKP-11 , cenerimod, amiselimod, CBP-307, OPL- 307, OPL-002, BMS-986166, SCD-044, BOS-173717, CP-1050, typically selected among mocravimod, FTY720 and siponimod, and most preferably mocravimod.
  • said S1 P receptor modulator is a S1 P receptor agonist. More preferably, the S1 P receptor modulator is of formula (I) or (II) : wherein
  • R 2 is H, halogen, trihalomethyl, C 1-4 alkoxy, C 1-7 alkyl, phenethyl or benzyloxy;
  • R 3 is H, halogen, CF 3 , OH, C 1-7 alkyl, C 1-4 alkoxy, benzyloxy, phenyl or C 1- 4 alkoxymethyl; each of R 4 and R 5 , independently is H or a residue of formula (a) wherein each of R 8 and R 9 , independently, is H or C 1-4 alkyl optionally substituted by halogen; and n is an integer from 1 to 4; and
  • R 6 is hydrogen, halogen, C 1-7 al kyl , C 1-4 alkoxy or trifluoromethyl, or pharmaceutically acceptable salts thereof, and more preferably mocravimod or pharmaceutically acceptable salts thereof.
  • the administration of the S1 P receptor modulator is started prior to allogenic HSCT, preferably 7, 8, 9, 10, 11 , 12, 13, or 14 days prior to HSCT, more preferably 11 days prior to HSCT.
  • the S1 P receptor modulator preferably mocravimod, is daily administered for a period set to at least 80 days, or at least 100 days or more, after the HSCT.
  • the S1 P receptor modulator is daily administered for at least 6, 7, 8, 9, 10, 11 , 12, 18, or 24 months, or more, preferably during the life of the subject or until relapse, i.e. chronic daily treatment.
  • the S1 P receptor modulator is daily administered from 1 to 14 days prior to HSCT, preferably from 11 days prior to HSCT, for at least 6, 7, 8, 9, 10, 11 , 12, 18, or 24 months, or more, or until relapse, after HSCT.
  • the amount of S1 P receptor modulator can be administered per day at a fixed amount.
  • said fixed daily dosage is 0,05 mg to 40 mg per day, preferably 0,1 mg to 35 mg, more preferably 0,5 mg to 30 mg, even more preferably 1 mg to 15 mg per day, even more preferably 1 ,5 mg to 7mg, even more preferably 2 mg to 5 mg, even more preferably about 3 mg per day.
  • the S1 P receptor modulator can be mocravimod and said mocravimod may be administered at a daily dose of 3mg, preferably three solid dosage form a day of 1 mg.
  • the conditioning regimen at step 2) includes a chemotherapy with a chemotherapeutic agent selected from the group consisting of cyclophosphamide or thiotepa, cytarabine, etoposide, busulfan or melphalan, fludarabine, and mixtures thereof such as a combined administration of fludarabine/busulfan, busulfan/cyclophosphamide, or fludarabine/melphalan.
  • a chemotherapeutic agent selected from the group consisting of cyclophosphamide or thiotepa, cytarabine, etoposide, busulfan or melphalan, fludarabine, and mixtures thereof such as a combined administration of fludarabine/busulfan, busulfan/cyclophosphamide, or fludarabine/melphalan.
  • the conditioning regimen is as follow:
  • the conditioning regimen at step 2) consists in : i. the administration of cyclophosphamide followed by total body irradiation, or ii. the administration of busulfan and cyclophosphamide, or ill. the administration of fludarabine and busulfan, and optionally, total body irradiation with a low dosage.
  • the hematopoietic stem cells are selected in step 3) from HLA-matched related or unrelated donor with 8/8 or higher matches at the HLA-A, -B, -C, -DRB1 , and/or -DQB1 loci, as determined by high resolution HLA typing.
  • the one or more immunosuppressants is selected from the group consisting of ciclosporin A, sirolimus, tacrolimus, methotrexate, and mycophenolate mofetil, preferably ciclosporin A or a mixture and methotrexate.
  • the immunosuppressant is at least ciclosporin A.
  • ciclosporin A can be administered within a period between the starting day of administration of the S1 P receptor modulator, preferably mocravimod, and the day of HSCT, preferably 3 days prior to HSCT.
  • the ciclosporin A is administered intravenously at an initial dosage of 2.5 mg/kg over 2 hours every 12 hours and optionally adjusted between 150 to 400 mg/L.
  • methotrexate is administered together with ciclosporin A the day of HSCT at a dosage of 10 mg/kg of and optionally 2 and 5 days after the first administration and the 16th day therefrom, at a dosage of 6 mg/kg.
  • the one or more immunosuppressants used in step 4) do not include tacrolimus.
  • the S1 P receptor modulator preferably mocravimod
  • said immunosuppressants treatment is reduced of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% compared to the starting dose of said immunosuppressant.
  • said subject is with refractory or relapsed AML after one or more AML therapy.
  • said subject is selected among the patients suffering of acute myelogenous leukemia and being either in first complete morphological remission but in the adverse-risk group called “CR1 high risk”, or in second and subsequent complete morphological remission called “CR2”, wherein said CR1 high risk, CR2 patients are defined according to ASBMT RFI 2017 - Disease Classifications Corresponding to CIBMTR Classifications of the American Society for Blood and Marrow Transplantation, preferably patients classified as CR1 high risk and CR2.
  • the subject is measurable residual disease (MRD)-positive prior to being administered the method of the present disclosure (e.g.HSCT with mocravimod).
  • the method prior to step (1 ), includes detecting the MRD status of a subject.
  • said S1 P receptor modulator is administered in an amount sufficient to treat AML and to prevent acute GVHD.
  • said S1 P receptor modulator is administered in an amount sufficient to treat AML and to prevent acute and chronic GVHD.
  • said patient is refractory GVHD-free, and relapse free at 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, or 24 months from HSCT.
  • said subject does not present one or more of the following:
  • GVHD grade lll/IV acute graft-versus-host disease
  • said methods and use described herein improves morbidity or mortality in a population of subjects, in particular via refractory GVHD-free, relapse- free survival (rGRFS) and both relapse-related mortality and transplant-related mortality at 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, 24 months from HSCT.
  • said methods and use reduce the occurrence, or severity of either GVHD, refractory GVHD, relapse or mortality during the next 12, 18 or 24 months after HSCT in a population of subjects.
  • said method improves the quality of life in a population of subjects, preferably as measured by the Fundation for the Accreditation of Cellular Therapy Bone Marrow Transplantation (FACT-BMT) questionnaire and/or the MD Anderson symptom inventory (MDASI), at 3, 6, 12 or 24 months as compared with the same method of HSCT without administration of said S1 PR modulator. Improvement may be significant, and for example the quality of life may increase of at least 10%, 20%, 30%; 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more.
  • FACT-BMT Fundation for the Accreditation of Cellular Therapy Bone Marrow Transplantation
  • MDASI MD Anderson symptom inventory
  • the disclosure relates to an S1 P receptor modulator, preferably mocravimod, for use in treating a human subject suffering from acute myelogenous leukemia (AML) and undergoing allogeneic hematopoietic stem cell transplant (HSCT).
  • AML acute myelogenous leukemia
  • HSCT allogeneic hematopoietic stem cell transplant
  • a “modulator” is a compound which, when administered to a subject, provides the desired interaction with the target receptor, either by way of the compound acting directly on the receptor itself, or by way of a metabolite of the compound acting on the receptor.
  • the S1 P receptor modulator preferably mocravimod, interacts with the S1 P receptor by either activating or inhibiting the receptor for signal transduction.
  • S1 P agonist refers to a compound which initiates a physiological response when combined with the S1 P receptor.
  • physiological response initiated is the agonist-induced internalization of the S1 P receptor.
  • the kinetics of agonist-induced internalization from cell membranes, and the recycling of the S1 P receptors to the cell membrane after said compound shedding depends on the compound.
  • S1 P receptor agonists may also be referred as functional antagonists.
  • the persistence of the internalization conditions the agonist's "functional antagonism" properties.
  • HSC refers to hematopoietic stem cell
  • HSCT refers to allogeneic hematopoietic stem cell transplantation.
  • pharmaceutically acceptable salts thereof includes both acid and base addition salts.
  • pharmaceutically acceptable acid addition salts include chlorides, hydrochlorides, bromides, sulfates, nitrates, phosphates, sulfonates, methane sulfonates, formates, tartrates, maleates, citrates, benzoates, salicylates, and ascorbates.
  • pharmaceutically acceptable base addition salts include sodium, potassium, lithium, ammonium (substituted and unsubstituted), calcium, magnesium, iron, zinc, copper, manganese, and aluminum salts.
  • Pharmaceutically acceptable salts may, for example, be obtained using standard procedures well known in the field of pharmaceuticals. For mocravimod, pharmaceutically acceptable salts would typically be acid-addition salts, since mocravimod is itself a base.
  • the pharmaceutically acceptable salts thereof is hydrochloride salt.
  • excipient refers to a non-active substance that is added alongside the drug substance, and is part of the formulation mixture.
  • Pharmaceutically acceptable excipient are for example fillers, solvents, diluents, carriers, auxiliaries, distributing and sensing agents, delivery agents, such as preserving agents, disintegrants, moisteners, emulsifiers, suspending agents, thickeners, sweeteners, flavoring agents, aromatizing agents, antibacterial agents, fungicides, lubricants, and prolonged delivery controllers, antioxidants, glidants.
  • delivery agents such as preserving agents, disintegrants, moisteners, emulsifiers, suspending agents, thickeners, sweeteners, flavoring agents, aromatizing agents, antibacterial agents, fungicides, lubricants, and prolonged delivery controllers, antioxidants, glidants.
  • the choice and suitable proportions of them are depended on the nature and way of administration and dosage.
  • an “average particle size” refers here to the D50 which means that 50% of the particles have size less than or equal to the indicated value.
  • an average particle size less than or equal to 8 pm refers to a D50 of 8 pm, i.e. 50% of the particles have a particle size less than or equal to 8 pm.
  • the term D90 means that 90% of the particles have a particle size less than or equal to the indicated value.
  • a D90 less than or equal to 25 pm means that 90% of the particles have a particle size less than or equal to 25 pm.
  • the D50 as well as the D90 are determined by laser light diffraction using the liquid route, e.g. on a BECKMAN-COULTER laser diffraction particle size analyzer LS 230, equipped with its small volume dispersion module (liquid route), following the technical manual and the manufacturer's instructions.
  • an effective amount refers to an amount of an active principle ingredient, for example, an S1 P receptor modulator, preferably mocravimod, that when administered to a subject, either as a single dose or as part of a series of doses, is effective to produce at least one therapeutic effect, either alone or in combination with other active agent.
  • an S1 P receptor modulator preferably mocravimod
  • patient refers to a human.
  • patient, subject or individual in need of treatment includes those who already have the disease, condition, or disorder, i.e. Acute Myelogenous Leukemia.
  • “Combination” refers to either a fixed combination in one dosage unit form, or a combined administration where a compound of the present disclosure and a combination partner (e.g. another drug as explained below, also referred to as “therapeutic agent” or “co-agent”) may be administered independently at the same time or separately within time intervals, especially where these time intervals allow that the combination partners show a cooperative, e.g. synergistic effect.
  • the single components may be packaged in a kit or separately.
  • One or both of the components e.g., powders or liquids
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • treating denotes reversing, alleviating, inhibiting the progress of, or preventing the disorder or condition to which such term applies, or reversing, alleviating, inhibiting the progress of, or preventing one or more symptoms of the disorder or condition to which such term applies.
  • AUCIast is defined as the concentration measured from time 0 to the last measurable concentration.
  • S1 P receptors are divided into five subtypes related G-coupled protein receptors (i.e., S1 Pi, S1 P2, S1 P3, S1 P4 and S1 P5), which are expressed in a wide variety of tissues and exhibit different cell specificity.
  • a modulator of the S1 P receptor for use according to the present methods of the disclosure is a compound which modulates one or more of the five S1 P receptor types 1 to 5 (SI PR1-5) by activating or inhibiting the receptor for signal transduction.
  • SI PR1-5 the five S1 P receptor types 1 to 5
  • Such compounds are also referred to herein as “S1 P agonists” and “S1 P inhibitor” respectively.
  • said S1 P receptor modulator for use in the treatment methods of the present disclosure is selected among KRP203 (mocravimod), FTY720 (fingolimod), siponimod (Mayzent®), fingolimod (GilenyaTM), ozanimod (Zeposia®), ponesimod, etrasimod, AKP-11 , cenerimod, amiselimod, CBP-307, OPL-307, OPL- 002, BMS-986166, SCD-044, BOS-173717, CP-1050.
  • said S1 P receptor modulator for use in the treatment methods of the present disclosure is selected among KRP203 (mocravimod), FTY720 (fingolimod), and Mayzent® (siponimod), most preferably mocravimod.
  • said S1 P receptor modulator for use in the treatment methods of the present disclosure is a S1 P agonist.
  • S1 P agonist is KRP203 (mocravimod), a SI PR1 selective agonist, or FTY720 (fingolimod), a multi-S1 PR agonist of S1 PR1, 3-5 or siponimod, a S1 PR 3 agonist, or any of their pharmaceutically acceptable salts.
  • said S1 P agonist is selected among those S1 P agonists activating selectively S1 PR1.
  • the S1 P receptor modulator for use according to the present disclosure is the compound of formula (I):
  • R 2 is H, halogen, trihalomethyl, C 1-4 alkoxy, C 1-7 alkyl, phenethyl or benzyloxy;
  • R 3 is H, halogen, CF 3 , OH, C 1-7 alkyl, C 1-4 alkoxy, benzyloxy, phenyl or C 1-4 alkoxymethyl; each of R 4 and R 5 , independently is H or a residue of formula (a) wherein each of R 8 and R 9 , independently, is H or C 1-4 alkyl optionally substituted by halogen; and n is an integer from 1 to 4; and
  • R 6 is hydrogen, halogen, C 1-7 al kyl , C 1-4 alkoxy or trifluoromethyl.
  • said compoud of formula (I) is an S1 P agonist, preferably an SI PR1 selective agonist.
  • R 3 is chlorine. More preferably, R 2 is H, R 3 is chlorine and R 6 is hydrogen.
  • R 2 is H, R 3 is chlorine, R 6 is hydrogen, and each of R 3 and R 5 , independently is H.
  • the S1 P receptor modulator for use according to the present disclosure preferably an S1 P agonist, preferably S1 PR1 selective agonist, is 2-amino-2- [4- (3-benzyloxyphenylthio) -2-chlorophenyl] ethyl-propane-1 ,3-diol, of formula (II) (also referred as mocravimod or KRP203): or pharmaceutically acceptable salts thereof.
  • S1 P receptor modulator for use according to the present disclosure, preferably S1 PR 1 selective agonist, includes the phosphate derivatives of the following formulae: Said compounds and their synthesis methods are also disclosed in W003/029205, W02004/074297, W02006/009092, W02006/041019 and WO2014128611A1 (which disclosures are incorporated herein by reference).
  • Mocravimod is particularly preferred. Indeed, comparing pharmacodynamic effects of different S1 P modulators, such as mocravimod, FTY720 and BAF312 established in healthy volunteers reveals differences in efficacy of lymphocyte sequestration.
  • the lymphocyte recovery time for KRP203 is significantly longer than for BAF312 and FTY720.
  • Particle size of the S1 P receptor modulator In the pharmaceutical industry, particle characterization of powder materials has become one of the crucial aspects in drug product development and quality control of solid oral dosage forms.
  • the particle size distribution of the drug substance may have significant effects on final drug product performance (e.g., dissolution, bioavailability, content uniformity, stability, etc.).
  • the particle size distributions of the drug substance can affect drug product manufacturability such as flowability, blend uniformity, compactibility, and have profound influence on almost every step of manufacturing processes for solid oral dosage forms, including pre-mixing/mixing, granulation, drying, milling, blending, coating, encapsulation, and compression.
  • the particle size of the drug substance can therefore ultimately impact safety, efficacy, and quality of the drug product.
  • the S1 P receptor modulator of the present disclosure has an average particle size (D50) of less than or equal to 8pm, preferably 6pm, more preferably 5pm. In another embodiment, the S1 P receptor modulator of the present disclosure has a D90 of less than or equal to 25pm, preferably 22pm, more preferably 19pm. Indeed, it has been found by the inventors that an average particle size (D50) of greater than 8 pm, preferably greater than 6pm, more preferably greater than 5pm and/or a D90 greater than 25 pm, preferably greater than 22pm, more preferably greater than 19pm, impairs the dissolution of the drug by diminishing the surface contact with the solvent resulting in lowering bioavailability and in vivo performance.
  • D50 average particle size
  • composition comprising the S1 P receptor modulator
  • the present disclosure also relates to a pharmaceutical composition of said S1 P receptor modulator, preferably mocravimod, as described above, in particular for their use in the treatment methods as disclosed.
  • the pharmaceutical composition of the present disclosure comprises the S1 P receptor modulator, preferably mocravimod, and one or more pharmaceutically acceptable excipients.
  • the pharmaceutical composition may be administered in any manner appropriate to the disease or disorder to be treated as determined by persons of ordinary skill in the medical arts.
  • An appropriate dose and a suitable duration and frequency of administration will be determined by such factors as discussed herein, including the condition of the patient, the type and severity of the patient’s disease, the particular form of the active ingredient, and the method of administration.
  • an appropriate dose (or effective dose) and treatment regimen provides the pharmaceutical composition in an amount sufficient to provide a therapeutic effect, for example, an improved clinical outcome, such as more frequent complete or partial remissions, or longer disease-free and/or overall survival, or a lessening of symptom severity or other benefit as described in detail herein.
  • compositions described herein may be administered to a subject in need thereof by any of several routes that can effectively deliver an effective amount of the compound.
  • the pharmaceutical composition may be administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically, bucally, or as an oral or nasal spray.
  • the pharmaceutical composition is suitable to be administered orally.
  • the pharmaceutical composition may be a solid dosage form suitable for oral administration.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the pharmaceutical composition is a capsule or a tablet.
  • the capsule may be a soft or hard gelatin capsule, preferably a hard gelatin capsule.
  • the capsule is HGC Crushed or HPMC capsules Crushed.
  • the release of the capsule or tablet content may be immediate or modified such as delayed, targeted or extended.
  • the solid dosage form is an immediate release dosage form.
  • the pharmaceutical composition comprises the S1 P receptor modulator, preferably mocravimod, and one or more pharmaceutically acceptable excipients, and particularly, at least one filler and mixtures thereof, a disintegrant, a lubricant and, a glidant.
  • Fillers are substance which are added to the drug substance in order to make the latter suitable for oral administration (e.g., capsules, tablets). Fillers themselves should not produce any pharmacological effect on human being. Examples of fillers include mannitol, microcrystalline cellulose, lactose monohydrate, anhydrous lactose, corn starch, xylitol, sorbitol, sucrose, dicalcium phosphate, maltodextrin, and gelatin.
  • the pharmaceutical composition of the present disclosure comprises at least one filler selected from mannitol, microcrystalline cellulose and mixtures thereof. In a preferred embodiment, the pharmaceutical composition of the present disclosure comprises a mixture of mannitol and microcrystalline cellulose.
  • Disintegrants are added to oral solid dosage forms to aid in their deaggregation. Disintegrants are formulated to cause a rapid break-up of solids dosage forms when they come into contact with moisture. Disintegration is typically viewed as the first step in the dissolution process.
  • disintegrants include the modified starch such as sodium starch glycolate, sodium carboxymethyl starch, and pre-gelatinized starch, crosslinked polymers, such as crosslinked polyvinylpyrrolidone (crospovidone) or crosslinked sodium carboxymethyl cellulose (croscarmellose sodium), and calcium silicate.
  • the pharmaceutical composition of the present disclosure comprises sodium starch glycolate as disintegrant.
  • Lubricants are substances that we use in tablet and capsule formulations in order to reduce the friction. Lubricant can facilitate extrusion of tablets from matrix, thus preventing formation of scratches on their surfaces.
  • lubricants can be divided into two groups: a) fats and fat-like substances; b) powdery substance. Powdery substances are more applicable then the fat-like ones, because the latter impact on solubility and chemical stability of the tablets. Powdery lubricants are introduced by powdering of granulate. They provide constant-rate outflow of mass for tabletizing from hopper into matrix that guaranties accuracy and constancy of the drug substance dosage.
  • lubricants examples include magnesium stearate, hydrogenated castor oil, glyceryl behenate, calcium stearate, zinc stearate, mineral oil, silicone fluid, sodium lauryl sulfate, L-leucine, and sodium stearyl fumarate.
  • the pharmaceutical composition of the present disclosure comprises magnesium stearate as lubricant.
  • Glidants are blended with the formulation to enhance the tablet-core blend-material flow property.
  • glidants are mixed within the particle arrangement of the tablet powder blend to improve flowability and uniformity within the die cavity of tablet presses. Glidants encourage the flow of tablet granulation by diminishing friction between particles. The effect of glidants on the flow of the granules depends on the size and shape of the particles of the granules and the glidants. Above a certain concentration, the glidant will in fact function to inhibit flowability.
  • glidants are usually added just prior to compression. Examples of glidants include colloidal silicon dioxide, starch, magnesium stearate and talc.
  • the pharmaceutical composition of the present disclosure comprises colloidal silicon dioxide as lubricant.
  • the pharmaceutical composition of the present disclosure comprises the S1 P receptor modulator, and at least one filler selected from mannitol, microcrystalline cellulose and mixtures thereof, sodium starch glycolate as disintegrant, magnesium stearate as lubricant and, colloidal silicon dioxide as glidant.
  • the dosage strength of the S1 P receptor modulator, preferably the hydrochloride salt of formula (I), in the solid dosage form is between 0.05 mg to 15 mg/unit, preferably between 0.1 mg to 10mg/unit, for example about 0.1 mg/unit, or about 0.4mg/unit, or about 1 mg/unit, or about 10 mg/unit, more preferably about 1 mg/unit.
  • composition of the present disclosure further comprises the following ingredients:
  • - mannitol preferably at a content from 48 to 88 mg/unit, more preferably from 58 to 78mg/unit, even more preferably at a content about 68 mg/unit;
  • microcrystalline cellulose preferably at a content from 5 to 45 mg/unit, more preferably from 15 to 35 mg/unit, even more preferably at a content about 25 mg/unit;
  • - sodium starch glycolate preferably at a content from 1 to 8 mg/unit, more preferably from 2 to 6 mg/unit, even more preferably at a content about 4 mg/unit;
  • - magnesium stearate preferably at a content from 0.025 to 4 mg/unit, more preferably from 0.5 to 2 mg/unit, even more preferably at a content about 1 mg/unit;
  • - colloidal silicon dioxide preferably at a content from 0.125 to 2 mg/unit, more preferably from 0.25 to 1 mg/unit, even more preferably at a content about 0.5 mg/unit.
  • composition of the present disclosure comprises the following ingredients:
  • the S1 P receptor modulator preferably mocravimod, preferably at a content from 0.05% to 15%, more preferably from 0.1 % to 10% mg/unit, even more preferably at a content about 0.1 % or 0.4% or 1 % or 10%;
  • - mannitol preferably at a content from 48% to 88%, more preferably from 58% to 78%, even more preferably at a content about 68%;
  • microcrystalline cellulose preferably at a content from 5% to 45%, more preferably from 15% to 35%, even more preferably at a content about 25%;
  • - sodium starch glycolate preferably at a content from 1% to 8%, more preferably from 2% to 6%, even more preferably at a content about 4%;
  • - magnesium stearate preferably at a content from 0.025% to 4%, more preferably from 0.5% to 2%, even more preferably at a content about 1 %;
  • - colloidal silicon dioxide preferably at a content from 0.125 to 2 mg/unit, more preferably from 0.25 to 1 mg/unit, even more preferably at a content about 0.5 mg/unit,
  • Percentage being expressed mg/mg of the total composition in dry weight.
  • the pharmaceutical composition of the disclosure is stable for at least 1 month at 50°C, preferably 2 months at 50°C.
  • the pharmaceutical composition of the disclosure is stable at least 24 months at 5°C. In another embodiment, the pharmaceutical composition of the disclosure is stable at least 24 months at 25°C/ 60% relative humidity.
  • stability of a composition is measured according to the following method: high-pressure liquid chromatography (HPLC) which is widely known in the field.
  • HPLC high-pressure liquid chromatography
  • a composition is stable if the sum of impurities is less than or equal to 0.7% with a confidence interval of [98-102]%.
  • Another aspect of the present disclosure relates to the process of preparing the above- mentioned pharmaceutical composition, wherein it comprises the step of: a. Blending the S1 P receptor modulator with microcrystalline cellulose, colloidal silicon dioxide, preferably at 22rpm for 18 mins, b. adding mannitol and blending the resulting mixture, preferably at 22rpm for 9 mins, c. adding sodium starch glycolate and blending the resulting mixture, preferably at 22rpm for 5 mins, d. adding magnesium stearate, blending the resulting pharmaceutical composition, preferably at 22rpm for 5 mins, e. recovering the pharmaceutical composition of the present disclosure.
  • the process further comprises the step of: f. filling the resulting pharmaceutical composition into capsules, g. recovering the resulting capsules filled with the pharmaceutical composition.
  • the patient population to be preferably targeted by the treatment methods
  • the treatment methods disclosed herein are suitable for patients having acute myeloid leukemia (AML) and that, as per standard medical practice, requires myeloablative conditioning followed by allogeneic hematopoietic stem cell transplant.
  • AML acute myeloid leukemia
  • the present method of treatments disclosed herein improves the GVL effect while reducing acute and chronic GVHD, and, optionally side effects of immunosuppressants, in particular calcineurin inhibitors, such as ciclosporin A, in particular for patients with high risk of relapse as defined below, and more specifically in combination with a chronic administration of S1 P receptor modulator and/or tapering of immunosuppressant treatment as described in the next sections.
  • immunosuppressants in particular calcineurin inhibitors, such as ciclosporin A
  • the methods of the present disclosure are particularly suitable for subject with refractory or relapsed AML after one or more AML therapy.
  • the term “refractory” means a subject which failed to achieve complete remission (e.g., wherein less than 5% of the cells in the bone marrow are blasts, and there is an absence of blasts with Auer rods in the bone marrow, an absence of extramedullary disease, and full hematologic recovery (e.g, absolute neutrophil count (ANC) >1 ,000/pL and platelet count >100,000/pL), and/or CRi following treatment for a disease, or achieved a CR e.g, wherein less than 5% of the cells in the bone marrow are blasts, and there is an absence of blasts with Auer rods in the bone marrow, an absence of extramedullary disease, and full hematologic recovery (e.g, absolute neutrophil count (ANC) >1 ,000/pL and platelet count >100,000/pL), and/or CRi lasting less than 90 days following treatment for the disease.
  • ANC absolute neutrophil count
  • ANC
  • relapse or “relapsed” has its ordinary meaning in the art, and may refer to the return of AML or the signs and symptoms of AML after a period of complete remission (e.g, initial complete remission) due to treatment.
  • relapse may refer to the recurrence of disease after complete remission meeting one or more of the following criteria (i) > 5% blasts in the marrow or peripheral blood, and/or (ii) extramedullary disease, and/or disease presence determined by a physician upon clinical assessment.
  • relapse refers to reoccurrence of a disease following a CR e.g, wherein less than 5% of the cells in the bone marrow are blasts, and there is an absence of blasts with Auer rods in the bone marrow, an absence of extramedullary disease, and full hematologic recovery (e.g, absolute neutrophil count (ANC) >1 ,000/pL and platelet count >100,000/pL), and/or CRi) lasting 90 days or longer.
  • ANC absolute neutrophil count
  • remission has its ordinary meaning in the art, and may refer to a decrease in or disappearance of signs and symptoms of cancer. In partial remission, some, but not all, signs and symptoms of cancer have disappeared. In complete remission (CR), all signs and symptoms of cancer have disappeared, although cancer still may be in the body. “Complete remission” as used herein, is measured by complete morphologic response (CMR). Complete morphological response is defined as leukaemia clearance ( ⁇ 5% marrow blasts and no circulating peripheral blasts) in conjunction with normal values for absolute neutrophil count and platelet count, no extramedullary manifestation of leukaemia and no need for repeat blood transfusions, and includes CR, CRi and CRh, as defined below.
  • complete remission of AML means the disease has been treated, and the following are true: (i) the complete blood count is normal; (ii) less than 5% of the cells in the bone marrow are blasts (leukemia cells); and (iii) there are no signs or symptoms of leukemia in the brain and spinal cord or elsewhere in the body.
  • complete remission of AML means less than 5% of the cells in the bone marrow are blasts, and there is an absence of blasts with Auer rods in the bone marrow, an absence of extramedullary disease, and full hematologic recovery (e.g ., absolute neutrophil count (ANC) >1 ,000/pL and platelet count >100,000/pL).
  • ANC absolute neutrophil count
  • CR as used herein, is defined as leukaemia clearance (less than 5% marrow blasts and no circulating peripheral blasts) in conjunction with normal values for absolute neutrophil count and platelet count, no extramedullary manifestation of leukaemia and no need for repeat blood transfusions.
  • CRi as used herein, is defined as meeting all CMR criteria except for an absolute neutrophil count ⁇ 1 ,000/ ⁇ L or platelet count ⁇ 100,000/ ⁇ L.
  • CRh as used herein, means less than 5% of the cells in the bone marrow are blasts, and there is an absence of blasts with Auer rods in the bone marrow, an absence of extramedullary disease, and partial hematologic recovery of both peripheral blood cell types (e.g, ANC >500/pL and platelet count >50,000/pL).
  • Partial remission means greater than or equal to 5% to less than or equal to 25% of the cells in the bone marrow are blasts, and a decrease of at least 50% in the percentage of blasts.
  • partial remission of AML means (i) greater than or equal to 5% to less than or equal to 25% of the cells in the bone marrow are blasts; (ii) a decrease of at least 50% in the percentage of blasts; and (iii) the complete blood count is normal.
  • the patient population is selected among the patients suffering from acute myeloid leukemia and classified as CR1 or CR2 according to CIBMTR classification.
  • said subject is selected among the patients suffering of acute myelogenous leukemia and being either in first complete morphological remission but in the adverse-risk group called “CR1 high risk”, or in second and subsequent complete morphological remission called “CR2”.
  • First complete remission or CR1 is defined according to “ASBMT RFI 2017 - Disease Classifications Corresponding to CIBMTR Classifications of the American Society for Blood and Marrow Transplantation” as patients having a treatment response, i.e. complete remission following the first treatment.
  • CR1 patients are classified in a 3-group classification (favorable, intermediate, adverse).
  • CR1 high risk patients corresponds to the adverse group as defined according to the 2017 European Leukaemia Network (ELN) genetic risk stratification (Dohner H, et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood. 2017 Jan 26;129(4):424- 447. doi: 10.1182/blood-2016-08-733196. Epub 2016 Nov 28.
  • EPN European Leukaemia Network
  • Second complete remission or CR2 is defined according to "ASBMT RFI 2017 - Disease Classifications Corresponding to CIBMTR Classifications of the American Society for Blood and Marrow Transplantation” (https://higherlogicdownload.s3.amazonaws.com/ASBMT/43a1f41f-55cb-4c97-9e78- c03e867db505/Uploadedlmages/ASBMT_RFI_2018B_CIBMTR_Disease_Classificati ons.pdf) as patients in complete remission (CR) for the second time and subsequent time.
  • CR2 patients achieved CR as defined above, relapsed and achieved at one complete remission again. Final pre-HSCT status must be complete remission.
  • CR2 includes CR2+ as defined according to "ASBMT RFI 2017 - Disease Classifications Corresponding to CIBMTR Classifications of the American Society for Blood and Marrow Transplantation”, i.e. patients in complete remission (CR) for more than the second time.
  • CR2+ patients achieved CR twice as defined above, relapsed and achieved complete remission again.
  • Stratification between CR1 and CR2 is needed for interpretation of the S1 P receptor modulator trial outcome, since HSCT basic performance may vary depending on CR status.
  • CR1 high risk, CR2 patients are male or female subjects aged 18- 70 years with acute myeloid leukaemia.
  • HSCT haematopoietic stem cell transplantation
  • the subject in need of the methods of treatment or use of the present disclosure is measurable residual disease (MRD)-positive prior to being administered the methods of treatment with HSCT and S1 P receptor modulator described herein (preferably , HSCT with mocravimod).
  • MRD residual disease
  • measurable residual disease and minimal residual disease refer to the post-therapy persistence of leukemic cells at levels below morphologic detection.
  • MRD is thought to be a strong prognostic indicator of increased risk of relapse or shorter survival in patients with hematologic cancers, such as AML.
  • MRD testing for AML is preferably conducted using one of three techniques: immunophenotypic detection by multiparameter flow cytometry (MFC), real-time quantitative PCR (RT- qPCR) and next-generation sequencing technology.
  • MFC uses panels of fluorochrome-labeled monoclonal antibodies to identify aberrantly expressed antigens of leukemic cells.
  • RT-qPCR can be used to amplify leukemia-associated genetic abnormalities.
  • Next-generation sequencing technology can be used to evaluate a few genes or an entire genome.
  • RT-qPCR and next-generation sequencing technology represent molecular approaches to MRD testing.
  • the European LeukemiaNet has issued consensus recommendations for the measurement of MRD in AML.
  • a percentage of cancer (e.g., AML) cells to leukocytes of 0.1 % or greater in a subject’s bone marrow, measured by MFC according to the ELN’s recommendations for MRD testing by MFC indicates the subject is MRD positive (MRD+) by MFC according to the ELN’s recommendations for MRD testing by MFC.
  • the ELN has also issued guidelines for molecular MRD testing in AML.
  • the ELN defines complete molecular remission as complete morphologic remission plus two successive negative MRD samples obtained within an interval of >4 weeks at a sensitivity level of at least 1 in 1 ,000, wherein the samples are collected and measured according to the ELN guidelines for molecular MRD testing.
  • the ELN defines molecular persistence at low copy numbers, which is associated with a low risk of relapse, as MRD with low copy numbers ( ⁇ 100-200 copies/104 ABL copies corresponding to ⁇ 1- 2% of target to reference gene or allele burden) in patients with morphologic CR, and a copy number or relative increase ⁇ 1 log between any two positive samples collected at the end of treatment, wherein the samples are collected and measured according to the ELN guidelines for molecular MRD testing.
  • the ELN defines molecular progression in patients with molecular persistence as an increase of MRD copy numbers >1 log 10 between any two positive samples collected and measured according to the ELN guidelines for molecular MRD testing.
  • the ELN defines molecular relapse as an increase of the MRD level of >1 log 10 between two positive samples in a patient who previously tested negative, wherein the samples are collected and measured according to the ELN guidelines for molecular MRD testing.
  • Both molecular persistence and molecular relapse are indicators of an MRD-positive subject by RT- qPCR conducted according to the ELN guidelines for MRD testing by RT-qPCR.
  • patients in complete molecular remission and patients labelled as having molecular persistence at low copy numbers are MRD-negative by RT-qPCR conducted according to the ELN guidelines for MRD testing by RT-qPCR.
  • RT-qPCR is the recommended molecular approach to MRD testing, as discussed in Ravandi, F., el al. and Schuurhuis, G. J., el al.
  • Specific recommendations for collecting and measuring samples (e.g . bone marrow samples) for MRD testing are described in Ravandi, F , et al, Blood Advances 12 June 2018, vol. 2, no. 11 and Schuurhuis, G. J., et al, Blood 2018 March 22, 131 (12): 1275-1291 , the relevant contents of which are incorporated herein by reference in their entireties.
  • the subject is MRD negative according to at least one of the ELN’s criteria described herein (e.g. MFC, molecular biology).
  • the subject is MRD- negative by MFC conducted according to ELN guidelines for MRD testing.
  • the subject is MRD-negative by RT-qPCR conducted according to ELN guidelines for MRD testing.
  • the subject is MRD-negative by both MFC and RT-qPCR conducted according to ELN guidelines for MRD testing. In some embodiments, the subject is MRD-negative by MFC conducted according to ELN guidelines for MRD testing, and is MRD-positive by RT-qPCR conducted according to ELN guidelines for MRD testing.
  • the subject is MRD-positive by MFC conducted according to ELN guidelines for MRD testing, and is MRD-negative by RT-qPCR conducted according to ELN guidelines for MRD testing.
  • MRD-negative according to one of the ELN’s criterion described herein e.g . the criterion for MFC
  • MRD-positive according to another of the ELN’s criterion described herein e.g. the criterion for RT-qPCR
  • a subject having a hematological cancer such as AML
  • a subject having a hematological cancer such as AML
  • the subject is MRD positive by the ELN’s criteria for MFC and RT-qPCR described herein.
  • a subject that is MRD positive for AML can be MRD-positive by MFC conducted according to ELN guidelines for MRD testing in AML, and MRD-positive by RT-qPCR conducted according to ELN guidelines for MRD testing in AML.
  • the method further comprises detecting the MRD status of a subject (e.g. prior to administering the HSCT methods of treatment with S1 P receptor modulator as described herein).
  • the S1 P receptor modulator as disclosed in the previous section, and more preferably mocravimod, and their pharmaceutical compositions, are useful as a drug in methods for treating AML in subjects undergoing HSCT, and more particularly in the subpopulation of patients as defined above.
  • the S1 P receptor modulator as disclosed in the previous section, and more preferably mocravimod, and their pharmaceutical compositions, are also useful as a drug in methods for preventing chronic GVHD in subjects undergoing HSCT, in particular in subjects undergoing HSCT for treating AML, and more particularly in the subpopulation of patients as defined above.
  • the method of the present disclosure comprises at least the following steps:
  • an S1 P receptor modulator for example, a compound of formula (I) or (II), or a pharmaceutically acceptable salt thereof;
  • conditioning said subject for destroying substantially the bone marrow and immune system wherein said conditioning includes treatment of said subject with an effective amount of a chemotherapeutic agent such as cyclophosphamide and/or treating said subject with a high-dose chemoradiation therapy;
  • a chemotherapeutic agent such as cyclophosphamide
  • an effective amount of S1 P receptor modulator (preferably mocravimod or other S1 P receptor modulator as disclosed above) is administered to the subject in need of such treatment.
  • an effective amount refers to the amount of S1 P receptor modulator (such as mocravimod) required to confer therapeutic effect on the subject, either alone or in combination with one or more other active agents. Effective amounts vary, as recognized by those skilled in the art, depending on, for example, route of administration, excipient usage, and co-usage with other active agents.
  • the desired therapeutic effect is inhibiting the progression of the disease. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods or can be monitored according to diagnostic methods discussed herein.
  • the desired response to treatment of the disease or condition also can be delaying the onset or even preventing the onset of the disease or condition.
  • the desired response can be delaying or preventing the onset of acute GVHD, chronic GVHD, relapse AML or death.
  • the desired response can be delaying or preventing the onset of AML relapse while delaying or preventing the onset of GVHD (grade lll/IV) refractory to systemic immunosuppressive treatment.
  • the desired response can be increasing survival at 3, 6, 12 and 24 months after HSCT.
  • the desired response can be improving the quality of life, in particular at 3, 6, 12 and 24 months after HSCT, as compared to same HSCT treatment without administration of S1 P receptor modulator.
  • the administration of the S1 P receptor modulator is started prior to allogenic hematopoietic stem cells transplantation (HSCT).
  • HSCT allogenic hematopoietic stem cells transplantation
  • administration is started at least 7 days prior to HSCT, for example 14, 13, 12, 11 , 10, 9, 8, or 7 days, preferably 11 days prior to HSCT.
  • the S1 P receptor modulator preferably mocravimod, is then daily administered for a period set to at least 80 days, or at least 100 days or more, after the HSCT.
  • the S1 P receptor modulator preferably mocravimod
  • the S1 P receptor modulator, preferably mocravimod is daily administered for a period between 6 and 24 months, for example for 12 months after the HSCT.
  • the daily dosage amount and dose of a S1 P receptor modulator, preferably mocravimod, described herein may depend upon the subject’s condition, that is, stage of the disease, severity of symptoms caused by the disease, general health status, as well as age, gender, and weight, and other factors apparent to a person of ordinary skill in the medical art.
  • the dose of the S1 P modulator, preferably mocravimod, for treating a human subject suffering from AML and undergoing allogeneic HSC transplant may be determined according to parameters understood by a person of ordinary skill in the medical art.
  • the amount of S1 P receptor modulator, preferably mocravimod, administered per day is a fixed amount.
  • the fixed daily dosage is 0,05 mg to 40 mg per day, preferably 0,1 mg to 35 mg, more preferably 0,5 mg to 30 mg, even more preferably 1 mg to 15 mg per day, even more preferably 1 ,5 mg to 7mg, even more preferably 2 mg to 5 mg, even more preferably about 3 mg per day.
  • the S1 P receptor modulator is mocravimod, and said mocravimod is daily administered for at least 6, 7, 8, 9, 10, 11 , 12, 18, 24 months, or more, after the HSCT at a dosage of 3 mg per day.
  • the daily dosage may be administered as one dose per day or in multiple doses in a single day. In a preferred embodiment, the daily dosage is administered once a day. In some embodiments, the doses are administered several times daily, preferably 3 times daily. The minimum dose that is enough to provide effective therapy may be used in some embodiments.
  • said S1 P receptor modulator is mocravimod, and said mocravimod is administered at a daily dose of 3mg, e.g. three solid dosage form a day of 1 mg.
  • 3 solid dosage form such as capsules or tablets, of 1 mg administered in a spaced-apart manner is easier to swallow than a single solid dosage form of 3 mg.
  • hematopoietic stem cell transplantation patients have to be conditioned by preferably using high dose of chemotherapy and/or performing total body irradiation (TBI) according to national guidelines adapted to institutional practices, also referred as chemoradiation therapy.
  • TBI total body irradiation
  • the standard of care is myeloablative regimen.
  • non myelo-ablative regimen or reduced intensity conditioning may also be used for some subset population of patients (Jethava et al., Bone Marrow Transplant. 2017 Nov;52(11 ):1504-1511 )
  • High-dose chemotherapy high-dose total body irradiation (TBI), or a combination thereof is carried out.
  • chemotherapeutic agent and “chemotherapy” refer to agents and therapies, respectively, that inhibit (e.g., arrest) the growth of cancer cells as, for example, by killing the cells or inhibiting cell division.
  • “Chemotherapeutic agent for AML” and “chemotherapy for AML” refer to chemotherapeutic agents and chemotherapies, respectively, administered to a subject with the purpose of treating AML in the subject.
  • chemotherapeutic agents examples include cyclophosphamide (CY), cytarabine (CA), etoposide (ETP), busulfan (BU), fludarabine (FLU), melphalan (MEL), methotrexate (MTX), ciclosporin A (CsA), and the like, and mixtures thereof such as fludarabine/busulfan, busulfan/cyclophosphamide and fludarabine/melphalan.
  • one of the following conditioning regimens may be administered (day numbers are relative to the day of HSCT).
  • conditioning regimens mention may be made of, for example:
  • conditioning regimens may occur to accommodate for patient’s condition and/or local practice. According to institutional practices, the 3 following conditioning regimens may be selected next to the standard regimen described hereabove:
  • a treatment consisting of administration of cyclophosphamide followed by total body irradiation a treatment consisting of administration of busulfan and cyclophosphamide and the like may be mentioned.
  • the allogeneic HSCT whose goal is two-fold: firstly, to replace the patient’s diseased hematopoietic system with new HSC stemming from a genetically disparate healthy donor, and secondly, to exploit the immunotherapeutic effect of the donor graft, i.e. graft-versus-leukaemia effect called “GVL”.
  • the allogenic HSCT may comprise transplanting HSCs from a donor to said patient. HSCs are collected from the donor and administered to the patient by intravenous infusion.
  • HSCs can be harvested from different sources. HSC mobilized by granulocyte colonystimulating factor (G-CSF) into the peripheral blood are the preferred source for transplantation today. Alternative sources for HSCs used clinically are bone marrow and umbilical cord blood. A suitable donor should be HLA-matched related or unrelated donor with 8/8 or higher matches at the HLA-A, -B, -C, -DRB1 , and/or -DQB1 loci, as determined by high resolution HLA typing.
  • G-CSF granulocyte colonystimulating factor
  • Allogeneic hematopoietic stem cell transplant may be performed as the standard of care. Suitable methods are described for example in Boglarka Gyurkocza, Andrew Rezvani & Rainer F Storb (2010) Allogeneic hematopoietic cell transplantation: the state of the art, Expert Review of Hematology, 3:3, 285-299, DOI: 10.1586/ehm.10.21.
  • stem cell source is mobilized peripheral blood collected via apheresis by a compatible donor.
  • the minimum recommended CD34+ cell dose in the graft may be 2 x 10 6 / kg, with a recommended target dose of about 5 x 10 6 / kg.
  • the S1 P receptor modulator preferably mocravimod, may also be used alone and/or in appropriate association, as well as in combination, with other pharmaceutically active compounds.
  • the S1 P receptor modulator preferably mocravimod
  • an immunosuppressive agent in particular during at least the first three months following HSCT.
  • immunosuppressive agents include without limitation ciclosporin A, sirolimus, tacrolimus, methotrexate, and mycophenolate.
  • the immunosuppressant agent comprises or essentially consists of an efficient amount of ciclosporin A.
  • Ciclosporin A or related immunosuppressant administration may be started, for example, within a period between the starting day of administration of the S1 P receptor modulator, preferably mocravimod, and the day of HSC transplant, preferably 3 or 2 days prior to HSC transplant or 1 day prior to HSC transplant.
  • intravenous administration of ciclosporin A may be carried out at an initial dosage of between 2 to 6 mg/kg/day, preferably 3 to 5 mg/kg/day. Dosage adjustments are carried out, based on the toxicity or the concentration of ciclosporin A relative to the target trough concentration (150 to 400 mg / L).
  • the administration of ciclosporin A can be changed to oral administration if the patient can tolerate the oral administration.
  • the initial dosage for oral administration may be set to the current dosage for intravenous administration.
  • the dosage of ciclosporin A is monitored at least weekly and changed to a clinically appropriate dosage.
  • the dosage level between 2 to 6 mg/kg/day, preferably 3 to 5 mg/kg/day may be continued during the day of HSC transplant for a period of up to 2 weeks before a change is made to oral maintenance therapy.
  • the daily doses of cyclosporin A may be about 12.5 mg/kg.
  • the maintenance therapy is continued for at least 3 months, preferably for 6 months before decreasing the dose.
  • the dose of cyclosporin A may be gradually decreased to zero by 1 year after transplantation.
  • the dosing schedule and the dosage of methotrexate will be adapted to the hospital standards.
  • HSCT e.g. 11 days after first administration of S1 PR modulator, preferably mocravimod
  • 10 mg/kg of methotrexate is administered, and 2 and 5 days after first administration and the 16th day therefrom, 6 mg/kg of methotrexate is administered, respectively.
  • the present methods advantageously enable to prevent acute and chronic GVHD and to preserve the graft versus leukemia effect while lowering immunosuppressor amount usually administered after an allogenic HSC transplantation, which thus lower adverse effects of such immunosuppressant treatments.
  • the immunosuppressants used in the method do not include tacrolimus.
  • Calcineurin inhibitors such as ciclosporin A (CsA) and tacrolimus (TAC) suppress donor T-cell activation and remain the most commonly used immunosuppressants for acute GVHD prophylaxis. These agents, however, also inhibit leukemia-specific T-cell responses leading to impaired GVL effect.
  • the chronic administration of an S1 P receptor modulator provides a significant prevention of chronic GVHD and enables to lower (taper) the amount of ciclosporin A or other immunosuppressive drugs to be administered to the subject in need thereof, thereby lowering the adverse effects of such immunosuppressive compound, while still preventing chronic GVHD and improving the GVL effect.
  • S1 P receptor preferably mocravimod
  • a long period for example at least 6 months, preferably 12, 18 or 24 months, typically during the life of the subject or until relapse
  • said subject is further treated with an efficient amount of immunosuppressants including at least ciclosporin A, and said immunosuppressants treatment is reduced or stopped prior to 6 months following HSCT, preferably, within a period from 3-6 months, or from 3-5 months, or from 3-4 months following HSCT.
  • said immunosuppressants treatment for example cyclosporine A treatment
  • said immunosuppressants treatment is reduced substantially (tapered), for example of at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% compared to the starting dose of said immunosuppressant.
  • Tapering of immunosuppressant treatment may be performed rapidly or gradually, for example with a gradual decrease of the dose for 1 , 2, 3, 4, 5, or 6 weeks after 1 , 2, or 3 months after HSC transplant, and/or discontinued treatment after at least 3, 4, 5, or 6 months after HSC transplant.
  • the immunosuppressive dose may be gradually reduced by 6 weeks to two months, and discontinued in 3 to 4 months after HSC transplant. The physician will determine the most appropriate tapering regimen depending on the conditions of the patient.
  • the method of the present disclosure comprises the administration of the S1 P receptor modulator, preferably mocravimod, in order to achieve: reduction in incidence of refractory GVHD, delay in relapse or increase in relapse-free survival, increase in overall survival or progression free survival, or improvement in quality of life.
  • the methods of the present disclosure delays or prevents the onset of acute GVHD, chronic GVHD, relapse AML or death, in particular for at least 3, 6, 9, 12, 18, or 24 months after HSCT.
  • the methods of the present disclosure delays or prevents the onset of AML relapse while delaying or preventing the onset of GVHD (grade lll/IV) refractory to systemic immunosuppressive treatment, in particular for at least 3, 6, 9, 12, 18, or 24 months after HSCT.
  • GVHD grade lll/IV
  • the methods reduce the morbidity or mortality in a population of subjects, in particular via GVHD-free, relapse-free, survival and both relapse-related mortality and transplant-related mortality.
  • said methods reduce the occurrence, or severity of either GVHD, refractory GVHD, relapse or mortality in particular for at least 3, 6, 9, 12, 18, or 24 months after HSCT, in a population of subjects.
  • said patient is refractory GvHD-free, relapse free (rGRFS) after at Ieast 3, 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, 18, 19, 20, 21 , 22, 23, or 24 months from HSCT.
  • rGRFS refractory GvHD-free, relapse free
  • rGRFS is calculated similarly to conventional GRFS treating grade III to IV acute GVHD, chronic GVHD requiring systemic treatment, and disease relapse/progression as events, except that GVHD that resolved and do not require systemic treatment at the last evaluation is excluded as an event in rGRFS.
  • said subject does not present one or more of the following:
  • GVHD grade lll/IV acute graft-versus-host disease
  • Also disclosed herein are methods for improving overall survival of subjects comprising for each subject treating the subject with any of the method using the S1 P receptor modulator as disclosed herein.
  • said method improves the quality of life of subjects, preferably as measured by the Foundation for the Accreditation of Cellular Therapy (FACT-BMT) Bone Marrow Transplantation questionnaire and/or the MD Anderson symptom inventory (MDASI), at 3, 6, 12 or 24 months as compared with subjects treated without S1 P receptor modulator administration according to the standard of care for HSCT. Improvement may be significant, and for example an increase of at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, or more.
  • the S1 P receptor modulator for use according to the present disclosure has
  • Cmax maximum plasma levels of between 0.4 ng/mL and 1 ng/mL, preferably between 0.55 ng/mL and 0.85 ng/mL, more preferably between 0.65 ng/mL and 0.75 ng/mL,
  • Tmax time to maximum plasma level
  • AUCIast area under the plasma concentration curve
  • the S1 P receptor modulator for use according to the present disclosure has
  • Cmax maximum plasma levels between 0.6 ng/mL and 1 .20 ng/mL, preferably between 0.75 ng/mL and 1 .05 ng/mL, more preferably between 0.85 ng/mL and 0.95 ng/mL,
  • Tmax time to maximum plasma level
  • the S1 P receptor modulator for use according to the present disclosure has a half time (T 1 ⁇ 2 ) of between 110h and 134h, preferably between 116h and 128h, more preferably between 121 h and 123h. In another embodiment, the S1 P receptor modulator for use according to the present disclosure has a half time (T V2) of between 100h and 122h, preferably between 105h and 117h, more preferably between 11 Oh and 112h.
  • Figure 1 Incidence of Mild chronic GVHD in patients who have received KRP 203 1 mg+ CsA, KRP203 3mg + CsA and KRP203 3 mg + TAC.
  • Figure 2 Favourable relapse incidence in patients who have received KRP 203 1 mg+ CsA, KRP203 3mg + CsA and KRP203 3 mg + TAC.
  • Figure 3 Incidence of Moderate chronic GVHD in patients who have received KRP 203 1 mg+ CsA, KRP203 3mg + CsA and KRP203 3 mg + TAC.
  • Example 1 HSCT with mocravimod for the treatment of AML
  • the therapy includes a daily chronic administration starting at day -11 prior to HSCT of an oral composition of mocravimod at a dose of 3mg per day, for a duration of at least 6 months, and preferably at least 12 months, or during the whole life of the subject (chronic administration), until relapse.
  • patients receive standard of care for GVHD prophylaxis starting preferably on day -4 prior allogeneic HSCT and which will include a combination of ciclosporin A and methotrexate.
  • the patient is subject to the following conditioning regimen: Fludarabine; 30 mg/m 2 i.v. once daily for 5 days on day -8 to -4 (150 mg/m2), Thiopeta; 5 mg/kg i.v. twice daily for 1 day on day -7 (10mg/kg) and Melphalan; 60 mg/m 2 i.v. once daily for 2 days on day -2 and -1 (120 mg/m 2 ).
  • Conditioning is followed by allogeneic HSCT.
  • G-CSF Granulocyte colony-stimulating factor
  • PBSC peripheral blood stem cells
  • Donor should be HLA- matched related or unrelated with 8/8 or higher matches at the HLA-A, -B, -C, -DRB1 , and/or -DQB1 loci, as determined by high resolution HLA typing.
  • Example 2 Clinical Study for treating AML patients with high risk of relapse
  • HSCT allogeneic hematopoietic stem cell transplantation
  • Mocravimod is a sphingosine 1 -phosphate receptor modulator (S1 P) developed by the applicant as an adjunctive therapy to allogeneic hematopoietic stem cell transplantation (allo-HSCT).
  • S1 P is a class of products associated with effect on the immune system. Some products are in development or are approved already as immunotherapy for the treatment of auto-immune diseases.
  • Fingolimod (Gilenya; FTY720) is approved for the treatment of multiple sclerosis (MS).
  • Mocravimod mechanism of action is similar to fingolimod. Both are prodrugs and their active phosphorylated metabolites block lymphocytes egress from lymph nodes thereby reducing the number of circulating lymphocytes.
  • mocravimod profile is attractive for being developed in conjunction with allo-HSCT and standard of care (SoC) to improve disease progression rate, overall survival and Quality of Life in patients with Acute Myelogenous Leukemia.
  • Treatment with mocravimod starting 11 days prior to and lasting up to 1 year post HSCT) will lead to sequestration of lymphocytes into secondary lymphoid organs.
  • the Treatment of the study is to be administered for the whole treatment phase (1 year) starting in the initial pre- transplantation period (11 days before HSCT) and in conjunction with a GVHD prophylaxis regimen based on CsA and MTX in the mocravimod arm, and according to the clinical site institutional practice in the control arm.
  • Allo-HSCT is a key component of the current standard of care in AML management and is indicated for induction failure (refractory), after Relapse, and for Consolidation of Complete Remission.
  • Allo-HSCT reduces the risk of relapse, but with a higher risk of treatment-related mortality.
  • allo-HSCT is indicated if the risk of relapse is > 35% (Dohner et al., Blood 2017 Jan 26;129(4):424-447) and if the patient’s age, comorbidities and frailty makes him fit for the procedure.
  • Allo-HSCT in High-risk AML is currently the only potential curative option. Although benefiting from GvL effect, this procedure is currently associated with a high risk of non-relapse morbidity and mortality due to GVHD.
  • the risk of GVHD is currently related to the intensity of the GvL effect and is mitigated by immunosuppressant prophylaxis, which in turn decreases the GvL effect.
  • Investigational treatment competing to HSCT in AML such as molecularly targeted drugs, monoclonal antibodies and immunoconjugates, Checkpoint Inhibitors, T-cells engagers and CAR-T cells may lead to durable remissions and thus may compete with HSCT through extending survival without the prospect of a cure.
  • Mocravimod’s potential to decrease GvH effect while preserving GvL is promising as a way to keep the prospect of a cure while decreasing transplant-related mortality and morbidity.
  • GVHD Graft vs Host Disease
  • GvL Graft vs Leukemia effect
  • GvHD may happen to be acute or chronic (aGvHD, cGvHD).
  • GvHD is common after alloHSCT with a high prevalence: aGvHD 30-70%, cGvHD 20- 50% (Magenau et al., Br J Haematol. 2016 Apr;173(2):190-205).
  • GvHD is a leading cause of post-transplant mortality and morbidity and a leading cause of post-transplant Quality of Life impairment.
  • GvHD is still an issue.
  • T-cell suppression via immunosuppressive prophylaxis by CNI and MTX, or first line therapy via systemic corticosteroids are associated with opportunistic infection, organ toxicity and risk of minimizing GvL effect.
  • manipulation of specific immune modulators can decrease effector T cells involved in GvHD, and GvL, while increasing regulatory T cells and downregulating inflammatory pathways.
  • Extra-Corporeal Photopheresis has been proposed as well in high risk/ Refractory aGvHD, possibly in combination with Mesenchymal Stem cells (MSC).
  • MSC Mesenchymal Stem cells
  • Fecal microbiota Transplant is investigative in aGvHD.
  • cGvHD management includes systemic corticosteroids with extended taper in first line, and ECP, rituximab or ibrutinib in second line
  • ST2 IL-1 R
  • REG3alpha intestinal injury biomarker
  • Post-transplant relapse in AML is biologically and clinically heterogenous. Some factors may impact the incidence of post-transplant relapse in AML.
  • GvHD prophylaxis intensive regimen based on Anti-Thymocyte Globulins (ATG) or ant-T cell antibodies, T cells depleted grafts are associated with high risk of relapse,
  • HSCT All patients beyond first relapse (CR2) are in need for HSCT and have a very high risk for relapse.
  • Clinical outcomes of HSCT vary depending on the depth of remission, and comorbidities, such as a fungal infection or organ dysfunction during induction.
  • HSCT outcome in CR2 AML is limited by the substantial number of patients who acquire comorbidities and cumulative chemotherapy toxicity compared to patients with HSCT in CR1.
  • Stratification CR2 vs CR1 is needed for interpretation of the mocravimod trial outcome, since HSCT basic performance may vary depending on CR status.
  • Post-transplant clonal profile often happens to be different from the clonal profile at diagnosis.
  • NGS tailored molecular monitoring
  • Multiparameter Flow Cytometry Multiparameter Flow Cytometry
  • routine Chimerism assessment to evaluate the MRD status in AML.
  • Small molecules such as FLT3-inhibitors, Hypo Methylating agents, or Prophylactic Donor Lymphocyte Infusions are maintenance therapies.
  • Post- transplant survival is mostly related to AML relapse after HSCT, but Non-Relapse mortality is significant.
  • Various strategies have been investigated to lower relapse and therefore improve OS.
  • dose intensity may depend on genomics.
  • Immune Checkpoint blockade CLA4, PD1 , PDL1 , etc..
  • targeted therapies TKIs
  • HMA maintenance hypomethylating agents
  • vaccines are investigational and may be associated with poor tolerability.
  • GRFS GVHD-free/relapse-free survival
  • events include grade 3-4 acute GVHD, systemic therapy-requiring chronic GVHD, relapse, or death in the first post- HSCT year.
  • rGRFS Refractory GRFS
  • rGRFS is calculated similarly to conventional GRFS treating grade III to IV acute GVHD, chronic GVHD requiring systemic treatment, and disease relapse/progression as events, except that GVHD that resolve and do not require systemic treatment at the last evaluation is excluded as an event in rGRFS.
  • rGRFS is more accurate than GRFS for Mocravimod assessment: death, relapse or persistent severe GVHD.
  • KRP203 is an immunomodulator that selectively targets sphingosine 1 - phosphate (S1 P) receptors via its phosphorylated active metabolite.
  • KRP203- phosphate is a potent agonist on human S1 P1 and S1 P4 and a potent but partial agonist on hS1 P3 and S1 P5. The therapeutic effects are believed to be due to the prevention of effector lymphocyte recirculation from lymphatic tissue to susceptible target organs such as the gut, skin or CNS.
  • KRP203-phosphate induces a long-lasting and complete internalization of the S1 P1 , which renders these cells unresponsive to S1 P, depriving them of an obligatory signal to egress from lymphoid organs.
  • KRP203 does not inhibit allogeneic-stimulated T cell proliferation in mixed lymphocyte reactions (MLR), exhibit hematological cytotoxicity nor induces apoptosis of CD4+ T cells at therapeutic concentrations in vitro.
  • KRP203 reduces lymphocyte counts in peripheral blood by more than 80%.
  • KRP203 did not show relevant potential in inhibiting the activity of human cytochrome P450 isoenzymes.
  • KRP203 and KRP203-phosphate did not show significant induction of CYP3A4 via the human PXR receptor up to concentrations of 6.15 pM and 50 pM, respectively.
  • KRP230 formed low amounts of covalent drug-protein adducts in human hepatocytes in vitro. Due to the low levels detected (68pmol/106 cells at 3 hours) in-vitro adduct formation is unlikely to be associated with clinical relevant findings at low dose levels (e.g. below 10 mg).
  • KRP203-phosphate/KRP203 ratios (based on AUC0-24h) of 1.4-6.6 and 10.9-19.1 were observed in dog and rat, respectively.
  • KRP203 was well tolerated up to high dose levels, producing lethality only at oral doses of 1000 mg/kg in rats or intravenous doses above 50 mg/kg in rats or mice. No mortality was seen following single oral dosing in dogs up to 2000 mg/kg.
  • mice Repeat-dose toxicity studies in mice (2 weeks) revealed mortality in this species at doses > 45 mg/kg/day, whilst in rats and dogs (up to 26 and 52-week, respectively) no mortality was seen up to daily doses of 50 mg/kg and 15 mg/kg, respectively.
  • effects related to lymphocyte depletion and characteristic changes in lymphoid organs were noted in all species at all dose levels tested in repeat dose toxicity studies. Besides these effects, the main target organs of toxicity were the lungs (mice, rats and dogs), the liver (mice and rats) and kidneys (mice).
  • the lungs were shown to be a consistent target organ in all animal species tested. At the end of the 13, 26 and 52-week rat and/or dog studies, no histopathological changes were observed at systemic exposures that were similar to those at which the lung effects occurred in the 4-week toxicity studies. Transitory activation of phagocytic cells and increased secretion of soluble markers by these cells is considered a main contributing factor to smooth muscle hypertrophy/hyperplasia.
  • hepatic effects were mild and did only occur at high dose levels in some studies in combination with general clinical signs (e.g., body weight and food consumption), indicating that the MTD may have been exceeded.
  • KRP203 does not show. in silico structural alerts for human liver side effects and genomic profiling of the liver in a 2-week exploratory toxicity study in rats did not indicate any hepatotoxicity-related changes.
  • the kidney was a target organ of toxicity in mice only and only in a 2-week study at doses > 45 mg/kg.
  • Minimal to severe lesions were noted in both sexes and consisted of tubular vacuolation, dilatation, degeneration, regeneration, hyaline droplets, hyaline casts, increase of mitotic figures, glomerular degeneration with fibrinoid deposits and/or glomerulopathy.
  • KRP203 causes marked eye irritation but is not irritating to rabbit skin and does not have a contact sensitizing potential.
  • Table 1 summarizes the clinical studies with the number of subjects exposed to KRP203.
  • Part 1 is a single arm open label study to investigate the safety of 3 mg/day KRP203 added to a standard of care GvHD prophylaxis (cyclosporine A (CsA)/methotrexate) in HSCT patients in 10 patients.
  • Part 2 is a randomized two arm open label study to compare the safety, efficacy and PK of 3 mg/day of KRP203 in combination with tacrolimus/methotrexate to 1 mg/day of KRP203 in combination with CsA/methotrexate in 20 patients.
  • CsA cyclosporine A
  • the study population comprised of male or female subjects who were diagnosed with a hematological malignancy and qualified for a standard allogeneic HSCT where HLA matched stem cell source was available.
  • malignancies included but were not limited to acute myeloid leukemia (AML), acute lymphocytic leukemia (ALL), myelodysplastic syndrome (MDS, chronic lymphocytic leukemia (CLL), marginal zone and follicular lymphomas, large-cell lymphoma, lymphoblastic, Burkitt’s and other high grade lymphomas; mantle-cell lymphoma, lymphoplasmacytic lymphoma; prolymphocytic leukemia or multiple myeloma.
  • AML acute myeloid leukemia
  • ALL acute lymphocytic leukemia
  • MDS myelodysplastic syndrome
  • CLL chronic lymphocytic leukemia
  • marginal zone and follicular lymphomas large-cell lymphoma, lymphoblastic, Burkit
  • Part 1 comprised of a screening period (Days -50 to -2), Baseline (Day -1 ), treatment period from Day 1 to Day 111 with KRP203 and a follow-up period up to 365 days (from transplant).
  • Subjects were admitted to the study site during the screening period (approximately 2 to 5 days before Baseline) and received the investigational product 3 mg KRP203 once daily from Day 1 until Day 111 along with cyclosporine A (CsA) given as standard of care (SOC) GvHD prophylaxis.
  • CsA cyclosporine A
  • SOC standard of care
  • Part 2 comprised a screening period (Days -50 to -2), treatment period from Day 1 to Day 111 with KRP203 and follow-up visits. Myeloablative conditioning was performed between Day 2 and Day 10 as per SOC using chemotherapy. Subjects who met the eligibility criteria were randomized to receive either:
  • AEs were related to system organ class gastrointestinal disorders (6, 60%) from start of treatment until just before transplant. From transplant up to 30 days of transplant, the most frequent system organ classes affected were gastrointestinal disorder (7, 70%), respiratory, thoracic and mediastinal disorders (4, 40%), vascular disorders (3, 30%), immune system disorders (3, 30%) and nervous system disorders (3, 30%). The most frequent AEs were pertaining to immune system disorders (10, 100%), general disorders and administration site conditions (6, 60%) and infection and infestations (5, 50%) after 30 days of transplant.
  • the top three system organ classes affected were gastrointestinal disorders, investigations and general disorder and administration site conditions. Up to 30 days after the transplant, the top three system organ classes reported were vascular disorders along with gastrointestinal disorders and investigations. After 30 days of the transplant, the majority (top three) of the AEs were pertaining to system organ class immune system disorders, infections and infestations and general disorders and administration conditions.
  • the most frequent AEs (at least in two subjects in either 1 mg KRP203 + CsA or 3 mg KRP203 + Tac treatment arm) reported from start of treatment until just before transplant were nausea, vomiting, weight increased, blood creatinine increased and diarrhea. Of these, the event of blood creatinine increased in one subject (1 mg KRP203 + CsA treatment arm) was suspected related to study treatment by the Investigator.
  • hepatic failure and Hodgkin’s disease were suspected to be related to study treatment by the Investigator.
  • an independent pathologist and the data monitoring committee also reviewed the case of hepatic failure and based on the histopathologic findings, concluded that the liver injury was rather caused by alloimmune-mediated hepatitis in line with acute GvHD (and thus, the hepatic failure was unlikely to be related to KRP203).
  • Hodgkin’s disease the Investigator suspected a relationship between Hodgkin's disease and KRP203. In the Investigator's opinion, KRP203 as an immunosuppression drug may have blunted the GVL effect which could have resulted in relapse of the underlying hematological disease.
  • HSCT For patients with high-risk Acute Myeloid Leukemia, an allogeneic HSCT remains the only curative option. This protocol offers these patients the opportunity to receive a promising adjunctive treatment, which improves Overall survival, and/or refractory GVHD-free, Relapse-free Survival (rGRFS) and/or Quality of Life associated to HSCT.
  • rGRFS refractory GVHD-free, Relapse-free Survival
  • the benefit expectations are based on previous Phase lib CKRP2032105 trial outcomes. As shown in Fig 1 , the incidence of mild chronic GVHD decreases in patients who have received 3 mg KRP203 and Ciclosporin A compared to patients who have received 3 mg KRP203 and Tacrolimus or 1 mg KRP203 and Ciclosporin A.
  • the objective of this study is to compare the safety and efficacy of mocravimod vs control as adjunctive treatment to allogeneic HSCT in subjects with Acute Myeloid Leukemia.
  • An additional objective of the study is to compare the effect of the two treatments on quality of life.
  • HSCT is performed with HLA- matched sibling or unrelated donor, in subjects with AML in first (CR1 ) or subsequent Complete Morphologic Remission (CR2).
  • rGRFS Refractory GVHD-free, relapse-free survival
  • GVHD grade lll/IV acute graft-versus-host disease
  • OS Overall survival
  • PFS Progression-free survival
  • Cumulative Incidence of Relapse is defined as incidence of morphologic or clinical relapse from the time of randomisation.
  • Quality of Life evaluation is conducted via questionnaire with primary endpoint at 12 months.
  • FACT-BMT Cellular Therapy- Bone Marrow Transplantation questionnaire
  • SF-36 Short Form 36-item health survey
  • MDASI MD Anderson Symptom Inventory
  • Randomization will use minimization to balance treatment groups with respect to Complete Remission status (CR1 vs CR2) and center.
  • a stochastic treatment allocation procedure will be used so that the treatment assignment is random for all subjects entered in the study.
  • Subjects randomized in the mocravimod group will receive 3 capsules dosed 1 mg once a day starting at Day-11 before the HSCT for a duration of 12 months or until all immunosuppressants have been tapered and weaning is completed for at least 4 weeks. Subjects who would have completed the 1 st year on treatment will be proposed to continue receiving study treatment for an additional year in an open label setting.
  • the study is designed to confirm the results of a non comparative Phase Ila clinical trial in hematologic malignancies and to compare the outcomes of patients receiving mocravimod as adjunctive therapy to HSCT to a control group of patients receiving a similar setting without mocravimod.
  • the study aims at showing superiority in the mocravimod group compared to the control group.
  • CR1 Complete remission
  • CR2 Complete remission
  • Subjects with a diagnosis of AML and related precursor neoplasms according to the WHO 2016 classification excluding acute promyelocytic leukaemia), including secondary AML after an antecedent haematological disease (e.g. myelodysplastic syndrome) and therapy-related AML
  • Subject is planned to undergo allogeneic HSCT from fully matched sibling donor or Unrelated Donor with an 8/8 match at HLA-A, -B, -C and DRB1 at high resolution by DNA-based typing, whereas Stem cell source is mobilized peripheral blood collected via apheresis by a compatible Donor.
  • the minimum recommended CD34+ cell dose in the graft should be 2 x 106/kg, and the recommended target dose should be 5 x 106/kg.
  • CR is defined as leukaemia clearance ( ⁇ 5% marrow blasts and no circulating peripheral blasts) in conjunction with normal values for absolute neutrophil count and platelet count, no extramedullary manifestation of leukaemia and no need for repeat blood transfusions.
  • CRi is defined as meeting all CMR criteria except for an absolute neutrophil count ⁇ 1 ,000/ ⁇ L or platelet count ⁇ 100,000/ ⁇ L.
  • a female is considered of childbearing potential following menarche and until becoming post-menopausal unless permanently sterile. Women are considered post-menopausal and not of child bearing potential if they have had 12 months of natural (spontaneous) amenorrhea with an appropriate clinical profile (e.g. age appropriate, history of vasomotor symptoms) or have had surgical bilateral oophorectomy (with or without hysterectomy) or tubal ligation at least six weeks ago. In the case of oophorectomy alone, only when the reproductive status of the woman has been confirmed by follow up hormone level assessment, she is considered not of child bearing potential.
  • Diagnosis of any previous or concomitant malignancy is an exclusion criterion, except when the subject completed treatment (chemotherapy and/or surgery and/or radiotherapy) with curative intent for this malignancy at least 6 months prior to enrolment
  • Concurrent severe and/or uncontrolled medical condition including o Clinically significant pulmonary fibrosis o Tuberculosis, except for history of successfully treated tuberculosis or history of prophylactic treatment after positive PPD skin reaction o Patients receiving chronic (daily) therapies for asthma o Patients with any other types of clinically significant bronchoconstructive disease o Uncontrolled diabetes mellitus as assessed by the investigator or diabetes complicated with organ involvement such as diabetic nephropathy or retinopathy. o Uncontrolled seizure disorder o Uncontrolled depression or history of suicide attempts/ideation
  • Cardiac dysfunction as defined by: o Myocardial infarction within the last 3 months of trial entry, or o Reduced left ventricular function with an ejection fraction ⁇ 40% as measured by multi-gated acquisition (MUGA) scan or echocardiogram (echo) within 28 days before screening, or o History or presence of stable or unstable ischemic heart disease (IHD), myocarditis, or cardiomyopathy, or o New York Heart Association (NYHA) Class ll-IV congestive heart failure, or o Unstable cardiac arrhythmias including history of or presence of symptomatic bradycardia, o Resting heart rate (physical exam or 12 lead ECG) ⁇ 60 bpm o History or current diagnose of ECG abnormalities indicating significant risk of safety such as: Concomitant clinically significant cardiac arrhythmias, e.g.
  • sustained ventricular tachycardia presence of a clinically relevant impairment of cardiac conduction including sick sinus syndrome, or sino-atrial heart block, clinically significant AV block, bundle branch block or resting QTc (Fridericia preferred, but Bazzet acceptable) > 450 msec for males and > 470 msec for females at Screening or Baseline ECG o History or presence of symptomatic arrhythmia or arrhythmia requiring treatment or being otherwise of clinical significance o Uncontrolled arterial hypertension; if controlled, the medication must be stable for three (3) months prior to baseline visit o Treatment with medication that impairs cardiac conduction (e.g., beta blockers, verapamil-type and diltiazem-type calcium channel blockers, or cardiac glycosides) o Concomitant use of agents known to prolong the QT interval unless they can be permanently discontinued for the duration of the study o Treatment with quinidine o History of syncope of suspected cardiac origin o History of familial long QT syndrome or known
  • Pulmonary dysfunction as defined by oxygen saturation ⁇ 90% on room air. Pulmonary function test (PFT) is required only in the case of symptomatic or prior known impairments within 28 days before screening - with pulmonary function ⁇ 50% corrected diffusing capacity of the lung for carbon monoxide (DLCO) and forced expiratory volume in 1 second (FEV1 )
  • HIV human immunodeficiency virus
  • HBV hepatitis B virus
  • HCV hepatitis C virus
  • NIMPs non-investigational medicinal products
  • the IMP is mocravimod.
  • NIMPS are products used in HSCT for conditioning, GVHD prophylaxis and infectious complications prophylaxis and are commercially available for human use under various brand names.
  • Mocravimod is formulated as a hard gelatin capsule and is an immediate release dosage form for oral administration.
  • the hard gelatin capsules contain white to off- white powder in a pink opaque (Swedish orange) capsule, size #4.
  • the hard gelatin capsule is administered at a regimen of a daily dose of 3 capsules by oral route starting at Day -11 before and for 1 year after HSCT.
  • traceability of mocravimod is defined as the ability to locate and identify each individual unit of mocravimod during any step from storage, to distribution to the subject or disposal. This also implies the ability to identify the subject at the trial site, and the ability to locate and identify all relevant data relating to products and materials coming into contact with the mocravimod. Accountability of the mocravimod will be documented at the trial centres. Detailed instructions on the accountability process are provided in the Pharmacy Manual.
  • Traceability of subjects is ensured by documenting the identity of subjects including their subject number at the trial site. Subject identities are protected and are only identified by code numbers that can be linked at the trial site to their full identity. Traceability of mocravimod batches from manufacturing until shipping to clinical sites is ensured by the procedures of the Contract Manufacturing Organization. Traceability of the mocravimod at the trial site is ensured by maintaining the mocravimod accountability log.
  • Mocravimod is prepared as hard gelatin capsules for oral administration as immediate release dosage form.
  • the capsules contain the following standard pharmacopoeial excipients: Mannitol; microcrystalline cellulose/cellulose, microcrystalline; sodium starch glycolate; magnesium stearate and colloidal silicon dioxide/silica; colloidal anhydrous.
  • the hard gelatin capsules contain white to off-white powder in a pink opaque (Swedish orange) capsule, size #4.
  • the packaging is HDPE bottles, each bottle contains 30 capsules, i.e. treatment for 10 days.
  • Mocravimod 1 mg hard gelatin capsules stored at 5°C/ambient RH is stable for 48 months when packaged in HDPE bottles and is stable for 24 months when packaged in PVC/PCTFE/PVC (Aclar) blister packs. Additionally, Mocravimod 1 mg hard gelatin capsules are stable for 36 months in HDPE bottles following storage at 25°C/60% RH. The drug product is also stable for 6 months when packaged in both HDPE bottles at 40°C/75% RH.
  • Subjects in the mocravimod arm will receive an immunosuppressive regimen for GVHD prophylaxis during approx. 6 months based on Ciclosporin A and Methotrexate. Tapering and weaning will be conducted according to the clinical site institutional practice.
  • CMV cytomegalovirus
  • EBV Epstein-Barr virus
  • patients who are EBV positive or have an EBV positive donor will be subject to regular quantitative PCR monitoring followed by adequate (pre-emptive) treatment if indicated. If quantified viral DNA levels exceed the institutional threshold for treatment of EBV (as established for each study center before participation in the study), patients should be treated with rituximab. It is also recommended to start rituximab if a patient who was EBV positive in the past, demonstrates enlarged lymph nodes, even if PCR for EBV is low or negative.
  • rituximab anti-CD20 375 mg/m2 IV is started once weekly, until PCR for EBV becomes negative.
  • PTLD PTLD is suspected on the basis of clinical symptoms
  • KRP203 is anticipated to be metabolized in the liver by CYP3A4 and 2D6 only to a limited extent (based on in vitro data), and the lack of effect of KRP203 and KRP203-P on cytochromes P450 enzymes and transporters, the risk of a PK DDI between KRP203 and CsA or TAC or other similar co-medications is considered low.
  • the weak inhibitory effect of ciclosporin on hepatic CYP3A4 is unlikely to relevantly impact the PK of KRP203.
  • the treatment phase corresponds to the start of the IMP regimen at Day -11 before HSCT in the mocravimod arm until the lastJMP intake.
  • the treatment phase in the control arm starts the day of the HSCT untill year post HSCT.
  • Enrolled subjects in the mocravimod arm will take 3 capsules on a daily basis for 1 year after HSCT.
  • a window of 7 additional days is tolerated in case the HSCT cannot happen as planned after conditioning for medical reasons (e.g. infections, AE monitoring).
  • the treatment phase will start in an outpatient setting followed rapidly by an inpatient (hospitalised) setting according to institutional practices for conditioning and HSCT. Subjects will be hospitalised from the start of the conditioning regimen until being discharged upon Investigator’s decision. Therefore, the treatment phase will be starting on Day -11 prior to HSCT and before conditioning to be administered on Days -6, -5, - 4, and -3 prior to HSCT.
  • Eligible subjects are randomized to either the mocravimod group or the control group. Subjects in both groups will undergo a conditioning regimen followed by HSCT as part of regular standard of clinical care. In this section the conditioning regimens and HSCT procedures are described.
  • conditioning regimens are to be administered (day numbers are relative to the day of HSCT). Scheduled deviations from these conditioning regimens are to be discussed between the principal investigator and the Medical Monitor of the Study.
  • rGRFS Disease Assessment rGRFS is a primary endpoint for efficacy assessment. Progression-Free Survival and Cumulative incidence of Relapse are secondary endpoints for efficacy assessment.
  • AML haematologic disease will be assessed at the local laboratory by BM biopsy or aspirate for morphology at the Screening Visit, Month 3, Month 6 and Month 12 Treatment Visits, Month 18 and 24 Follow-up Visits, unless relapse has already been confirmed. AML disease assessment will also be performed in case of suspected relapse.
  • a morphologic relapse is defined as morphological evidence of leukemia in Bone Marrow, or at other extra-medullary sites.
  • a bone marrow biopsy cannot be obtained, it may be replaced by a bone marrow aspirate. If a bone marrow aspirate and/or biopsy had already been obtained within 6 weeks prior to a scheduled visit, the assessment does not need to be repeated.
  • GVHD events will be diagnosed and classified based on the NIH criteria (Filipovich et al., Biol Blood Marrow Transplant. 2005 Dec;11 (12):945-56; Jagasia et al., Biol Blood Marrow Transplant 2015 Mar;21 (3):389-401 .e1 ) as summarized in Table 3.
  • Table 3 Categories of acute and chronic GVHD are summarized in Table 3.
  • Acute GVHD will be graded according to the modified Glucksberg scale (Harris et al., Biol Blood Marrow Transplant 2016 Jan;22(1 ):4-10) (see below).
  • Chronic GVHD will be graded according to NIH criteria (Filipovich et al., Biol Blood Marrow Transplant. 2005 Dec;11 (12):945-56; Jagasia et al., Biol Blood Marrow Transplant 2015
  • the start date of the GVHD event is defined as the date of initiation of GVHD treatment or the date of biopsy confirmation of GVHD, whichever is earlier.
  • Resolution of the GVHD event is defined as complete response, i.e. resolution of all signs and symptoms.
  • TRM Transplant-related mortality
  • FACT-BMT The Foundation for the Accreditation of Cellular Therapy- Bone Marrow Transplantation questionnaire (FACT-BMT, version 4 and the MD Anderson Symptom Inventory (MDASI) will be scored at Screening, Day+14, Day + 28, Day + 60, Month 3, Month 6, Month 12, and Month 24, provided that validated translations in local language are available.
  • MDASI MD Anderson Symptom Inventory
  • SAP statistical analysis plan
  • null and alternative hypotheses are:
  • rGRFS On the primary endpoint, rGRFS, it is anticipated that the 12-month rGRFS will be at least 60% in the mocravimod arm versus 40% at most in the control group (a hazard ratio equal to 0.55). A power of 0.8 will be required for this magnitude of treatment effect, and the two-tailed significance level will be set at 0.05.
  • the sample size required is 140 subjects, and 116 events are required for the final analysis of rGRFS to take place. To compensate for potential drop off and losses to follow-up at one year, a total of 160 subjects will be enrolled into this study.
  • the ITT population consists of all randomized patients.
  • the ITT population is the primary efficacy dataset for the primary and secondary endpoints.
  • the MITT population consists of all randomized patients who received an HSCT and mocravimod (active group) or at least one dose of GVHD prophylaxis (control group).
  • Per protocol (PR) population Prior to locking the database, the sponsor will define a PP population as a subset of the MITT population of patients without major protocol deviations.
  • the PP population will be used to (1 ) shed light on potential reasons why the primary analysis of the ITT population may have failed to reach significance, or (2) investigate how major protocol deviations may have had an impact on the magnitude of the treatment benefit.
  • the definition of “major” protocol deviations will be agreed upon, and all cases of such major deviations adjudicated prior to database lock, by a team blinded to treatment allocation.
  • the safety population consists of all patients who received an HSCT.
  • treatment groups will be compared through the Cochran-Mantel- Haenszel test, stratified by CR1 vs CR2. The impact of prognostic factors upon these endpoints will be assessed through logistic regression models.
  • descriptive statistics (cumulative incidences or proportion of patients free of the event of interest) will be presented for time to event endpoints at 3 months, 6 months, 12 months and 24 months.
  • the 95% confidence intervals will be calculated for the treatment contrast.
  • the primary endpoint of the study is rGRFS, treated as a time to event variable.
  • the primary approach for the primary efficacy analysis will be a randomization test to reflect the treatment allocation procedure.
  • the randomization test will be based on a large number of simulated trials, say S, in which the treatments are re-allocated to the patients actually entered in the study (in the same order of entry) using the same minimization algorithm.
  • Each simulated trial uses a different seed for the random number generator.
  • the significance probability (P-value) of the randomization test is calculated directly from the empirical distribution of A under the null hypothesis.
  • s be the number of ⁇ l_’s for which
  • the two- sided randomization P-value is equal to s/S.
  • the number of simulated trials S will be chosen to ensure that the P-value is estimated correctly to the second significant digit (Buyse M., Stat Med. 2010 Dec 30;29(30):3245-57).
  • AML Acute myeloid leukaemia
  • AML (megakaryoblastic) with t(1 ;22)(p13.3;q13.3);RBM15-MKL1
  • Provisional entity AML with BCR-ABL1
  • Provisional entity AML with mutated RUNX1
  • TAM Transient abnormal myelopoiesis
  • DRI Disease Risk Index
  • the DRI is in the public domain and therefore available for public use (Armand P., et al. Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation. Blood. 2014; 123(23): 3664-3671 ).
  • ALL Acute lymphoblastic leukaemia
  • AML cytogenetics Favorable: lnv(16); Intermediate: Normal, All other abn.; Adverse: Complex (>4 abn.)
  • EBMT European Group for Blood and Marrow Transplantation
  • the EMBT risk score is in the public domain and therefore available for public use (Gratmple A. The EBMT risk score. Bone Marrow Transplant. 2012; 47(6):749-56).
  • Table C The EBMT Risk Score 1 1 The EBMT risk score is calculated as the sum of the scores for each of the five risk factors
  • the Glucksberg acute GVHD score is in the public domain and therefore available for public use (Gratmple A. The EBMT risk score. Bone Marrow Transplant. 2012; 47(6):749-56).
  • Stage 1 maculopapular rash involving ⁇ 25% of the body surface
  • Stage 2 maculopapular rash involving 25-50% of the body surface
  • Stage 3 maculopapular rash involving > 50% of the body surface
  • Stage 4 generalised erythroderma (> 50% of the body surface) plus bullous formation and desquamation (> 5% of the body surface)
  • Stage 0 no or intermittent nausea, vomiting, or anorexia
  • Stage 1 persistent nausea, vomiting or anorexia
  • Stage 0 ⁇ 500 mL/day or ⁇ 3 episodes/day
  • Stage 1 500-999 mL/day or 3-4 episodes/day
  • Stage 2 1000-1500 mL/day or 5-7 episodes/day
  • Stage 3 > 1500 mL/day or > 7 episodes/day
  • Stage 4 severe abdominal pain with or without ileus or grossly bloody stool (regardless of stool volume)
  • Stage 0-3 Stage 2-3 Stage 2-3 Stage 0-1 and/or
  • NASH National Institutes of Health
  • the NIH chronic GVHD score is in the public domain and therefore available for public use (Filipovich et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report. Biol Blood Marrow Transplant. 2005;11 (12):945-56; Jagasia et al. National Institutes of Health consensus development project on criteria for clinical trials in chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report. Biol Blood Marrow Transplant. 2015;21 (3):389-401 ).
  • the NIH global scoring system for chronic GVHD reflects the clinical effect of chronic GVHD on the patient’s functional status. Eight organs or sites (skin, mouth, eyes, gastrointestinal tract, liver, lungs, joint and fascia, and genital tract) are considered for calculating global score. Elements included in the proposed global scoring include both the number of organs or sites involved and the severity score within each affected organ. Performance status scoring is not incorporated into the global scoring system. The global descriptions of mild, moderate, and severe were chosen to reflect the degree of organ impact and functional impairment due to chronic GVHD.
  • the global scoring system can be applied only after the diagnosis of chronic GVHD is confirmed by either (1 ) the presence of a diagnostic feature or, if a diagnostic feature is not present, (2) at least 1 distinctive manifestation of chronic GVHD with the diagnosis supported by histologic, radiologic, or laboratory evidence of GVHD from any site.
  • Moderate chronic GVHD o 3 or more organs involved with no more than score 1 OR o at least 1 organ (not lung) with a score of 2 OR o lung score 1
  • Severe chronic GVHD o At least 1 organ with a score of 3 OR o lung score of 2 or 3
  • mice undergo conditioning with 7-9 Gy of total body irradiation (TBI), which is followed by intravenous injection of 5-10x10 6 T-cell depleted bone marrow (TCDBM) supplemented with 1 -2.5x10 7 splenocytes from allogeneic B10.D2 donors to induce chronic GVHD.
  • TBI total body irradiation
  • TDBM T-cell depleted bone marrow
  • No GVHD controls receive donor cells from syngeneic Balb/c mice.
  • Some recipients in the GVHD group receive 3 mg/d KRP203 starting on day -1 (i.e., one day prior to bone marrow transplant (BMT) and chronic GVHD induction on day 0) until final analysis.
  • Chronic GVHD is evaluated at different time points post-BMT up to day 42.
  • Classical pathological changes of chronic skin GVHD e.g., reduced hair follicles and fibrosis, is analyzed in harvested skin samples.
  • the effect of KRP203 on chronic GVHD is confirmed in a second murine model of chronic GVHD.
  • donor / recipient combination is either LP/J / C57BL/6 or C57BL/6 / B10.BR, for analysis of chronic GVHD with or without skin involvement.
  • KRP203 administration start on day -1 until final analysis. The earliest final analysis is on day 28 post BMT. Analysis time points are up to day 47 post BMT. In all models, analyses include histology of target organs (e.g., skin, kidney, lung) including pathology scoring, pulmonary function test and immune cell phenotyping.
  • Example 4 Preparation of pharmaceutical composition comprising the S1P receptor modulator 12 excipients have been selected for the compatibility study of KRP203 (the S1P receptor modulator). In order to select the optimal combination of excipients and considering the risk of loss of stability, the following 17 blends have been prepared according to the table 4 below. Table 4 s euspa c
  • Example 5 Stability data of the pharmaceutical compositions of example 1 stored at 50°C
  • the 17 blends of example 1 have been prepared in both dry and wet condition and stored at defined conditions for 6 weeks.
  • the 17 blends were stored at defined conditions for 3, 6 weeks and were analyzed for the assay and the degradation products. Table 5
  • blends 7, 10, 16 and 17 were found to be highly compatible with assay data of 98.7%, 98.4%, 99,5% and 98.4% respectively at the end of 6 weeks.
  • the blend 17 was found to be the optimal blend from the selection as it showed the best stability while including the required excipient for a capsule or tablet formulation of the S1 P receptor modulator, namely, a filler, a disintegrant, a lubricant, and a glidant.
  • Example 6 The pharmaceutical composition according to the invention.
  • VSE add the first sub-lot of microcrystalline cellulose, KRP203 hydrochloride and the colloidal silicon dioxide to a grey polypropylene container and blend in the Turbula mixer for 3 mins at 24 rpm.
  • a capsule filling speed challenge will be performed on the MG2 Labby and captured in the development record. • Encapsulate the blend (100mg fill weight) into capsules (size 4) using the MG2 Labby Encapsulation machine. Perform in-process weight checks every 20 minutes on 20 capsules.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Emergency Medicine (AREA)
  • Transplantation (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
PCT/IB2021/000033 2021-01-28 2021-01-28 Methods of treatment with s1p receptor modulators WO2022162409A1 (en)

Priority Applications (16)

Application Number Priority Date Filing Date Title
AU2021424131A AU2021424131A1 (en) 2021-01-28 2021-01-28 Methods of treatment with s1p receptor modulators
PCT/IB2021/000033 WO2022162409A1 (en) 2021-01-28 2021-01-28 Methods of treatment with s1p receptor modulators
MX2023008907A MX2023008907A (es) 2021-01-28 2022-01-27 Metodos de tratamiento con moduladores del receptor de esfingosina-1-fosfato (s1p).
JP2023545210A JP2024506825A (ja) 2021-01-28 2022-01-27 S1p受容体調節剤による処置の方法
KR1020237025402A KR20230136603A (ko) 2021-01-28 2022-01-27 S1p 수용체 조정제를 사용한 치료 방법
CA3208122A CA3208122A1 (en) 2021-01-28 2022-01-27 Methods of treatment with s1p receptor modulators
EP23191736.0A EP4248958A3 (en) 2021-01-28 2022-01-27 Methods of treatment with s1p receptor modulators
PCT/EP2022/051940 WO2022162088A2 (en) 2021-01-28 2022-01-27 Methods of treatment with s1p receptor modulators
AU2022214268A AU2022214268A1 (en) 2021-01-28 2022-01-27 Methods of treatment with s1p receptor modulators
TW111103784A TW202245741A (zh) 2021-01-28 2022-01-27 使用s1p受體調節劑之治療方法
EP22702938.6A EP4284348A2 (en) 2021-01-28 2022-01-27 Methods of treatment with s1p receptor modulators
CN202280012135.2A CN116723835A (zh) 2021-01-28 2022-01-27 使用s1p受体调节剂的治疗方法
US18/066,883 US20230130403A1 (en) 2021-01-28 2022-12-15 Methods of treatment with s1p receptor modulators
IL304729A IL304729A (en) 2021-01-28 2023-07-25 Treatment methods with s1p receptor modulators
CL2023002212A CL2023002212A1 (es) 2021-01-28 2023-07-26 Métodos de tratamiento con moduladores del receptor de esfingosina-1-fosfato (s1p).
CONC2023/0009893A CO2023009893A2 (es) 2021-01-28 2023-07-26 Métodos de tratamiento con moduladores del receptor de esfingosina-1-fosfato (s1p)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/IB2021/000033 WO2022162409A1 (en) 2021-01-28 2021-01-28 Methods of treatment with s1p receptor modulators

Publications (1)

Publication Number Publication Date
WO2022162409A1 true WO2022162409A1 (en) 2022-08-04

Family

ID=74701516

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/000033 WO2022162409A1 (en) 2021-01-28 2021-01-28 Methods of treatment with s1p receptor modulators

Country Status (3)

Country Link
CN (1) CN116723835A (zh)
AU (1) AU2021424131A1 (zh)
WO (1) WO2022162409A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4282407A1 (en) * 2022-05-27 2023-11-29 Priothera SAS Treatment of cancer with s1p receptor agonists

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029205A1 (fr) 2001-09-27 2003-04-10 Kyorin Pharmaceutical Co., Ltd. Derive de sulfure de diaryle, son sel d'addition et immunosuppresseur
WO2004074297A1 (ja) 2003-02-18 2004-09-02 Kyorin Pharmaceutical Co., Ltd. アミノホスホン酸誘導体とその付加塩及びs1p受容体調節剤
WO2006009092A1 (ja) 2004-07-16 2006-01-26 Kyorin Pharmaceutical Co., Ltd. 効果的な医薬の使用法及び副作用発現の防御に関する方法
WO2006041019A1 (ja) 2004-10-12 2006-04-20 Kyorin Pharmaceutical Co., Ltd. 2-アミノ-2-[2-[4-(3-ベンジルオキシフェニルチオ)-2-クロロフェニル]エチル]-1,3-プロパンジオール塩酸塩又はその水和物の製造方法及びその製造中間体
WO2014128611A1 (en) 2013-02-20 2014-08-28 Novartis Ag Treatment of graft versus host disease in transplant patients
WO2020022507A1 (ja) 2018-07-27 2020-01-30 杏林製薬株式会社 造血幹細胞移植を受けた患者における血液悪性腫瘍の再発抑制剤

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003029205A1 (fr) 2001-09-27 2003-04-10 Kyorin Pharmaceutical Co., Ltd. Derive de sulfure de diaryle, son sel d'addition et immunosuppresseur
WO2004074297A1 (ja) 2003-02-18 2004-09-02 Kyorin Pharmaceutical Co., Ltd. アミノホスホン酸誘導体とその付加塩及びs1p受容体調節剤
WO2006009092A1 (ja) 2004-07-16 2006-01-26 Kyorin Pharmaceutical Co., Ltd. 効果的な医薬の使用法及び副作用発現の防御に関する方法
WO2006041019A1 (ja) 2004-10-12 2006-04-20 Kyorin Pharmaceutical Co., Ltd. 2-アミノ-2-[2-[4-(3-ベンジルオキシフェニルチオ)-2-クロロフェニル]エチル]-1,3-プロパンジオール塩酸塩又はその水和物の製造方法及びその製造中間体
WO2014128611A1 (en) 2013-02-20 2014-08-28 Novartis Ag Treatment of graft versus host disease in transplant patients
WO2020022507A1 (ja) 2018-07-27 2020-01-30 杏林製薬株式会社 造血幹細胞移植を受けた患者における血液悪性腫瘍の再発抑制剤

Non-Patent Citations (27)

* Cited by examiner, † Cited by third party
Title
ABO-ZENAHORWITZ, CURR OPIN PHARMACOL, vol. 2, no. 4, August 2002 (2002-08-01), pages 452 - 7
ARBER DA, SEMIN HEMATOL, vol. 56, no. 2, April 2019 (2019-04-01), pages 90 - 95
ARMAND P. ET AL.: "Validation and refinement of the Disease Risk Index for allogeneic stem cell transplantation", BLOOD, vol. 123, no. 23, 2014, pages 3664 - 3671
BOGLARKA GYURKOCZAANDREW REZVANIRAINER F STORB: "Allogeneic hematopoietic cell transplantation: the state of the art", EXPERT REVIEW OF HEMATOLOGY, vol. 3, no. 3, 2010, pages 285 - 299
BUYSE M., STAT MED., vol. 29, no. 30, 30 December 2010 (2010-12-30), pages 3245 - 57
DERTSCHNIG S ET AL., BLOOD, vol. 122, no. 5, 2013, pages 837 - 841
DERTSCHNIG S ET AL., BLOOD, vol. 125, no. 17, 2015, pages 2720 - 2723
DOHNER H ET AL.: "Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel", BLOOD, vol. 129, no. 4, 26 January 2017 (2017-01-26), pages 424 - 447, XP055754595, DOI: 10.1182/blood-2016-08-733196
FILIPOVICH ET AL., BIOL BLOOD MARROW TRANSPLANT, vol. 11, no. 12, December 2005 (2005-12-01), pages 945 - 56
FILIPOVICH ET AL.: "National Institutes of Health consensus development project on criteria for clinical trials in chronic graft-versus-host disease: I. Diagnosis and staging working group report", BIOL BLOOD MARROW TRANSPLANT., vol. 11, no. 12, 2005, pages 945 - 56, XP025312589, DOI: 10.1016/j.bbmt.2005.09.004
GRATWOHL A.: "The EBMT risk score", BONE MARROW TRANSPLANT., vol. 47, no. 6, 2012, pages 749 - 56
HARRIS ET AL., BIOL BLOOD MARROW TRANSPLANT, vol. 22, no. 1, January 2016 (2016-01-01), pages 4 - 10
HASHIMOTO D ET AL., EUR J IMMUNOL., vol. 37, 2007, pages 271 - 281
HOLTAN ET AL., BIOL BLOOD MARROW TRANSPLANT, vol. 21, no. 6, June 2015 (2015-06-01), pages 1029 - 36
HUU DOANH LE ET AL: "FTY720 Ameliorates Murine Sclerodermatous Chronic Graft-Versus-Host Disease by Promoting Expansion of Splenic Regulatory Cells and Inhibiting Immune Cell Infiltration Into Skin : FTY720 Ameliorates Murine Sclerodermatous Chronic GVHD", ARTHRITIS & RHEUMATISM, vol. 65, no. 6, 30 May 2013 (2013-05-30), US, pages 1624 - 1635, XP055843797, ISSN: 0004-3591, DOI: 10.1002/art.37933 *
JAGASIA ET AL., BIOL BLOOD MARROW TRANSPLANT, vol. 21, no. 3, March 2015 (2015-03-01), pages 389 - 401
JAGASIA ET AL., BLOOD, vol. 119, no. 1, 5 January 2012 (2012-01-05), pages 296 - 307
JAGASIA ET AL.: "National Institutes of Health consensus development project on criteria for clinical trials in chronic Graft-versus-Host Disease: I. The 2014 Diagnosis and Staging Working Group report", BIOL BLOOD MARROW TRANSPLANT., vol. 21, no. 3, 2015, pages 389 - 401
JETHAVA ET AL., BONE MARROW TRANSPLANT., vol. 52, no. 11, November 2017 (2017-11-01), pages 1504 - 1511
KIM ET AL., J CLIN INVEST., vol. 111, 2003, pages 659 - 669
MAGENAU ET AL., BR J HAEMATOL., vol. 173, no. 2, April 2016 (2016-04-01), pages 190 - 205
RAVANDI, F. ET AL., BLOOD ADVANCES, vol. 2, no. 11, 12 June 2018 (2018-06-12)
RUGGERI ET AL., J HEMATOL ONCOL., vol. 9, no. 1, 17 September 2016 (2016-09-17), pages 89
SCHUURHUIS, G. J. ET AL., BLOOD, vol. 131, no. 12, 22 March 2018 (2018-03-22), pages 1275 - 1291
STORB ET AL., N ENGL J MED, vol. 314, no. 12, 20 March 1986 (1986-03-20), pages 729 - 35
YOKOYAMA E ET AL., BONE MARROW TRANSPLANT., vol. 55, 2020, pages 787 - 795
YOKOYAMA EMI ET AL: "Short-term KRP203 and posttransplant cyclophosphamide for graft-versus-host disease prophylaxis", BONE MARROW TRANSPLANTATION, NATURE PUBLISHING GROUP, GB, vol. 55, no. 4, 4 November 2019 (2019-11-04), pages 787 - 795, XP037079719, ISSN: 0268-3369, [retrieved on 20191104], DOI: 10.1038/S41409-019-0733-8 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4282407A1 (en) * 2022-05-27 2023-11-29 Priothera SAS Treatment of cancer with s1p receptor agonists
WO2023227760A1 (en) * 2022-05-27 2023-11-30 Priothera Sas Treatment of cancer with s1p receptor agonists

Also Published As

Publication number Publication date
CN116723835A (zh) 2023-09-08
AU2021424131A1 (en) 2023-07-27

Similar Documents

Publication Publication Date Title
JP7445893B2 (ja) ヒトにおける固形腫瘍の処置のためのC.novyi
JP6906564B2 (ja) 一次癌療法後に使用するためのホウ素含有プロテアソーム阻害剤
EP3986403A1 (en) Azacitidine in combination with venetoclax, gilteritinib, midostaurin or other compounds for treating leukemia or myelodysplastic syndrome
Martín et al. Lenalidomide in combination with R‐ESHAP in patients with relapsed or refractory diffuse large B‐cell lymphoma: a phase 1b study from GELTAMO group
Munakata et al. Phase 1 study of tazemetostat in Japanese patients with relapsed or refractory B‐cell lymphoma
CN113194952A (zh) Hdm2-p53相互作用抑制剂和bcl2抑制剂的组合及其治疗癌症的用途
US20230130403A1 (en) Methods of treatment with s1p receptor modulators
CN113412262A (zh) 包含杂环蛋白激酶抑制剂的制剂
JP2023080357A (ja) リンパ球悪性疾患を治療するための方法
CN114302746A (zh) 包含抗cd25抗体药物缀合物和另一剂的组合疗法
WO2022162409A1 (en) Methods of treatment with s1p receptor modulators
TW202011948A (zh) 接受造血幹細胞移植的患者之血液惡性腫瘤的復發抑制劑
US20100226917A1 (en) Methods for the treatment of hematologic malignancies
CN115279347A (zh) Ahr抑制剂和其用途
JP2018502863A (ja) 白血病の治療のための4−(4−フルオロ−2−メトキシフェニル)−n−{3−[(s−メチルスルホンイミドイル)メチル]フェニル}−1,3,5−トリアジン−2−アミンの使用
WO2020068754A1 (en) Methods of treating myeloproliferative disorders
US20180021276A1 (en) Bezafibrate for the treatment of cancer
EP3265075B1 (en) Roneparstat combined therapy of multiple myeloma
JP2018509440A (ja) リンパ腫の治療のための4−(4−フルオロ−2−メトキシフェニル)−n−{3−[(s−メチルスルホンイミドイル)メチル]フェニル}−1,3,5−トリアジン−2−アミンの使用
AU2022407330A1 (en) Combination therapy comprising a pkc inhibitor and a c-met inhibitor
CN115768480A (zh) 使用蛋白激酶c(pkc)抑制剂和细胞毒性剂的治疗

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21707771

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021424131

Country of ref document: AU

Date of ref document: 20210128

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21707771

Country of ref document: EP

Kind code of ref document: A1