WO2017142331A1 - Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance - Google Patents

Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance Download PDF

Info

Publication number
WO2017142331A1
WO2017142331A1 PCT/KR2017/001726 KR2017001726W WO2017142331A1 WO 2017142331 A1 WO2017142331 A1 WO 2017142331A1 KR 2017001726 W KR2017001726 W KR 2017001726W WO 2017142331 A1 WO2017142331 A1 WO 2017142331A1
Authority
WO
WIPO (PCT)
Prior art keywords
dosage
growth hormone
patient
administration
week
Prior art date
Application number
PCT/KR2017/001726
Other languages
English (en)
Inventor
Tae Kyung Kim
Young_Joo AHN
Jung Won Woo
Mi Soo KANG
Sang-In YANG
Ji-Young Seo
Hyun Hee Park
Ji-Eun CHA
Mi Sun Byun
Joan Yoon Ji LEE
Original Assignee
Genexine, Inc.
Handok Inc.
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
Priority to CN201780018326.9A priority Critical patent/CN109152816B/zh
Priority to JP2018543675A priority patent/JP6896749B2/ja
Priority to RU2018132694A priority patent/RU2732113C2/ru
Priority to CA3014164A priority patent/CA3014164A1/fr
Priority to PL17753492.2T priority patent/PL3416677T3/pl
Priority to DK17753492.2T priority patent/DK3416677T3/da
Application filed by Genexine, Inc., Handok Inc. filed Critical Genexine, Inc.
Priority to BR112018016817A priority patent/BR112018016817A2/pt
Priority to ES17753492T priority patent/ES2925901T3/es
Priority to US16/077,177 priority patent/US20210177945A1/en
Priority to EP17753492.2A priority patent/EP3416677B1/fr
Priority claimed from KR1020170021104A external-priority patent/KR20170096968A/ko
Publication of WO2017142331A1 publication Critical patent/WO2017142331A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner

Definitions

  • the present disclosure relates to a pharmaceutical composition for treating growth hormone deficiency, containing hGH-hyFc (GX-H9) which is a recombinant human growth hormone prepared by fusing hybrid Fc to human growth hormone hGH. More particularly, the present disclosure relates to a appropriate method of administering a recombinant hGH which is effective to treat the growth hormone deficiency and to a pharmaceutical composition for treating growth hormone deficiency, including a recombinant hGH GX-H9 and a pharmaceutically acceptable carrier, in which the recombinant hGH GX-H9 is administered once a week with a dosage of 0.1 to 0.3 mg per weight kg of a patient or twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient.
  • GX-H9 hGH-hyFc
  • the present disclosure relates to a method for treating growth hormone deficiency, including administering an recombinant hGH GX-H9 to a patient with growth hormone deficiency once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient or twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient.
  • Growth hormone is a hormone which is secreted from an anterior pituitary gland, as a single molecular polypeptide composed of 191 amino acids. Insulin like growth factor-1 (IGF-1) is expressed in combination with a growth hormone receptor to be involved in growth and regeneration of cells. It is known that the growth hormone is produced in the pituitary gland of the normal human body, and gradually increased until adolescence and gradually decreased with age.
  • IGF-1 Insulin like growth factor-1
  • the most common growth hormone deficiency is adult growth hormone deficiency (AGHD) and pediatric growth hormone deficiency (PGHD).
  • AGHD adult growth hormone deficiency
  • PGHD pediatric growth hormone deficiency
  • the adult growth hormone deficiency occurs when the patient’s pituitary gland is injured by radiation or surgery in the treatment of brain tumors and cerebral hemorrhage, or idiopathically. If the secretion of the growth hormone is not performed well, symptoms including a loss in weight, reduction of the mineral density of the bone, increase in the fat, reduction of HDL, increase of LDL, decrease in muscle strength, and the like are shown, and thus, life quality is deteriorated.
  • the concentration of IGF-1 in serum belongs to a standard deviation score (SDS) of -2 or less ( ⁇ -2 SDS) or within 2.5 percentile ( ⁇ 2.5 percentile) as compared with normal people in the same age group.
  • SDS standard deviation score
  • the response value of the growth hormone in the blood may be measured by stimulation tests such as an insulin tolerance test (ITT), an arginine load test (GHRH + ARG), a glucagon test, an L-DOPA test and a clonidine test.
  • the peak GH of the growth hormone is 11.0 ⁇ g/L or less in patients with a body mass index (BMI) of less than 25 kg/m 2 , 8.0 ⁇ g/L or less in patients with a BMI of 25 to 30 kg/m 2 , or 4.0 ⁇ g/L or less in patients with a BMI of more than 30 kg/m 2 , the peak GH is considered as deficiency (Guidelines for Use of Growth Hormone in Clinical Practice, Endocr. Pract . 2009; 15 (Suppl 2)).
  • a growth hormone secretion disorder occurs when damage of the pituitary gland or developmental disorders is present.
  • a growth hormone secretion disorder is shown as short stature, and height with growth lower 3% or 5 cm or less a year in the growth curve of the same age, and symptoms such as hypoglycemia, deterioration of stamina, depression, and mental immaturity may be shown.
  • the symptom may be determined as pediatric growth hormone deficiency (Consensus guideline for the diagnosis and treatment of GH deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society, J. Clin . Endocrinol. Metab ., 2000 Nov; 85(11): 3990-3).
  • the dosage of the drug was selected on the basis of the weight of the patient in the related art, but recently, the patient is treated with the individualized dosage. That is, the treatment starts at a dosage lower than an expected therapeutic optimum dosage and adjusted by a method of increasing or decreasing 0.1 to 0.2 mg/day dosage depending on clinical response, adverse events (fasting glucose), or a IGF-1 level. Gender, estrogen status, and age of the patient need to be considered when selecting the treatment dosage of the growth hormone.
  • the goal of treatment in patients with adult growth hormone deficiency is to improve metabolism normalization and quality of life.
  • the IGF-1 level in the blood needs to be optimized to the middle (50th percentile or 0 SDS) to 1 SDS of the normal range (-2 SDS to 2 SDS) of dosage according to age and gender.
  • the pediatric growth hormone deficiency it is recommended that treatment starts as soon as possible after being diagnosed as a patient.
  • a method of subcutaneous administration of growth hormone every night is used and the recommended dosage is 25 to 50 ⁇ g/kg/day.
  • the growth rate is checked periodically at 3 months or 6 months and it is recommended to verify adverse events for verifying height growth, a change in growth rate, patient’s individual compliance, and safety, and to verify IGF-1 or IGFBP-3 levels in the serum.
  • a treatment goal in patients with pediatric growth hormone deficiency is to normally grow the height and growth hormone needs to be administered so that the IGF-1 levels in the blood are close to the average of same age (50th percentile or 0 SDS).
  • Growth hormone is extracted from the pituitary gland of dead body when the growth hormone treatment starts for the first time in the 1950s, and the supply is very limited and the cost is expensive because the amount of growth hormone extracted from one person is very small.
  • the recombinant growth hormone drugs currently marketed in the USA include Genotropin by Pfizer, Humatrope by Eli Lilly, Nutropin by Genentech, Norditropin by Novo Nordisk, and the like.
  • the recombinant growth hormone preparations are daily formulations requiring administration 6 times or 7 times a week.
  • Humatrope is used with a dosage of 0.2 mg/day (a range of 0.15 to 0.30 mg/day).
  • the starting dosage is 0.2 mg/day (a range of 0.15 to 0.3 mg/day) and can be changed to a dosage of 0.1 to 0.2 mg/day in 1 to 2 month cycles.
  • the dosage of Nutropin is set based on the body weight, the starting dosage is used so as not to exceed 0.005 mg/kg/day or more.
  • the dosage is increased so as not to exceed 0.01 mg/kg/day after 4 weeks of administration.
  • Norditropin is not based on body weight and set as a dosage
  • the starting dosage is 0.2 mg/day (a range of 0.15 to 0.3 mg/day) and can be changed to a dosage of 0.1 and 0.2 mg/day in 1 to 2 month cycles.
  • the dosage of Norditropin is set based on the body weight, the starting dosage is used so as not to exceed 0.004 mg/kg/day or more. If it is necessary to increase the dosage, the dosage is increased so as not to exceed 0.016 mg/kg/day or more after 6 weeks of administration.
  • Genotropin is used with a dosage of 0.16 to 0.24 mg/kg/week and Humatrope is used with a dosage of 0.026 to 0.043 mg/kg/day.
  • Nutropin is used with a dosage of 0.3 mg/kg/week and Norditropin is used with a dosage of 0.024 to 0.034 mg/kg/day.
  • growth hormone preparations are one-day formulations, and particularly, in the case of pediatric patients, it is inconvenient to inject the drug every day for a long treatment period of 3 to 4 years, and it is known that the mental stress caused by injection reduces the quality of life of the patient. In addition, dosing compliance of frequency by patient becomes the biggest factor that hinders the treatment effect. Further, it is also known that the number of administration failures increases markedly as increasing treatment duration ( Endocrine practice , 2008 Mar; 14 (2): 143-54). Approximately 2/3 of patients have low compliance according a default, and actually, it is known to reduce a height growth speed ( PloS one , 2011 Jan; 6(1): e16223).
  • Nutropin depot was developed by Genentech in the US as a monthly formulation, but due to is difficulty of production, it was withdrawn from the market.
  • Eutropin Plus/Declage by LG Life Sciences developed a weekly formulation using hyaluronic acid (HA), but due to needle larger needle size complared to first generation, it contains inconvenience.
  • GX-H9 hGH-hybrid Fc
  • hGH-hybrid Fc hGH-hybrid Fc
  • US Patent Registration No. 7,867,491 a hybrid type Fc capable of overcoming complement dependent cytotoxicity and antibody dependent cellular cytotoxicity which are problems of the existing Fc fusion technology was prepared by combining immunoglobulin IgD and immunoglobulin IgG4. Subsequently, in US Patent Registration No.
  • hGH-hyFc a recombinant hGH (hGH-hyFc, GX-H9) which is a material capable of replacing an existing daily-type growth hormone formulation
  • hGH-hyFc a recombinant hGH
  • GX-H9 a recombinant hGH which is a material capable of replacing an existing daily-type growth hormone formulation
  • the actual half-life in the body and therapeutic dosage of the Fc fused protein is greatly changed depending on what kind of pharmacologically active ingredient is bound to Fc.
  • the effective and safe dosages and its frequency for the treatment of growth hormone deficiency using GX-H9, in which human growth hormone hGH is fused to hyFc, have not yet been found.
  • An object of the present disclosure is to providing a method for treating growth hormone deficiency comprising administering recombinant hGH, GX-H9, to a patient with growth hormone deficiency once with an interval of at least a week and with a dosage of at least 0.1 mg per weight kg of a patient.
  • An object of the present disclosure is to providing a method for treating growth hormone deficiency using an recombinant hGH GX-H9 by determining a dosage and a dose frequency of the recombinant hGH GX-H9 which is effective to treat the growth hormone deficiency.
  • An aspect of the present disclosure provides a pharmaceutical composition for treating growth hormone deficiency comprising a recombinant human growth hormone, GX-H9, and a pharmaceutically acceptable carrier, wherein the recombinant hGH is administered once with an interval of at least a week and with a dosage of at least 0.1 mg per weight kg of a patient.
  • An aspect of the present disclosure provides a pharmaceutical composition for treating growth hormone deficiency comprising a recombinant human growth hormone, GX-H9, and a pharmaceutically acceptable carrier, in which the recombinant hGH is administered once a week with a dosage of 0.1 to 0.3 mg per weight kg of a patient.
  • Another aspect of the present disclosure provides a pharmaceutical composition for treating growth hormone deficiency comprising an recombinant hGH GX-H9 and a pharmaceutically acceptable carrier, in which the recombinant hGH is administered twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of a patient.
  • Yet another aspect of the present disclosure provides a method for treating growth hormone deficiency including administering recombinant human growth hormone, GX-H9, to a patient with growth hormone deficiency once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient.
  • Still another aspect of the present disclosure provides a method for treating growth hormone deficiency including administering a recombinant human growth hormone, GX-H9, to a patient with growth hormone deficiency twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient.
  • the recombinant human growth hormone, GX-H9 when administered to a patient with growth hormone deficiency once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient or twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient, the level of growth hormone in the body can be maintained for a longer period and in respect to hGH level, IGF-1 SDS value may be maintained in normal range for longer time.
  • giving possibility to treat growth hormone deficiency by administering a growth hormone once a week or twice-monthly without the need for daily administration.
  • FIG. 1 illustrates a result of binding affinity of a Fc ⁇ receptor Fc ⁇ R I for the recombinant hGH (GX-H9);
  • FIG. 2 illustrates a result of binding affinity of C1q for the recombinant hGH (GX-H9)
  • FIG. 3 is a result of the weight gain in hypophysectomized rats
  • FIG. 4 illustrates charactieristics of pharmacodynamics in a single subcutaneous administration of the recombimant hGH (GX-H9) to rats;
  • FIG. 5 illustrates the characteristics of pharmacodynamics in a single subcutaneous administration of the recombinant hGH (GX-H9) in monkeys;
  • FIG. 6 illustrates the charactreisicis of pharmacodynamics in a repeated subcutaneous administration of the recombinant hGH (GX-H9) in monkeys;
  • FIG. 7 illustrates the characteristics of pharmacodynamics of the recombinant hGH (GX-H9) in a Phase 1 clinical study
  • FIG. 8 illustrates the characterisitics of pharmacokinetic (IGF-1 SDS) of the recombinant hGH (GX-H9) in a Phase 1 clinical study;
  • FIG. 9 illustrates the characteristics of pharmacodynamics in a repeated administration of the recombinant hGH (GX-H9) in a Phase 2) clinical study.
  • FIG. 10 illustrates the characteristics of pharmacokinetic in a repeated administration of the recombinant hGH (GX-H9) in a Phase 2 clinical study.
  • the inventors conducted clinical studies in order to develop the dosage and the dose frequency capable of exhibiting an optimal effect of GX-H9, by targeting 32 healthy adults (2013-002771-18) and 45 adult patients with growth hormone deficiency (2014-002698-13, EudraCT).
  • the recombinant hGH, GX-H9 once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient or twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient, the growth hormone in the body is maintained for a longer period and the IGF-1 SDS value may be maintained in normal range.
  • One aspect of the present disclosure relates to pharmaceutical composition for treating growth hormone deficiency containing a recombinant hGH (GX-H9) and a pharmaceutically acceptable carrier, wherein the recombinant hGH is administered once with an interval of at least a week and with a dosage of at least 0.1 mg per weight kg of a patient.
  • GX-H9 recombinant hGH
  • One aspect of the present disclosure relates to pharmaceutical composition for treating growth hormone deficiency containing a recombinant hGH (GX-H9) and a pharmaceutically acceptable carrier, in which the recombinant hGH is administered once a week with a dosage of 0.1 to 0.3 mg per weight kg of a patient.
  • the present disclosure relates to a pharmaceutical composition, in which the recombinant hGH is administered once a week with a dosage of 0.1 to 0.2 mg per weight kg of a patient.
  • another aspect of the present disclosure relates to a pharmaceutical composition for treating growth hormone deficiency containing an recombinant hGH protein GX-H9 and a pharmaceutically acceptable carrier, in which the recombinant hGH is administered twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of a patient.
  • the present disclosure relates to a pharmaceutical composition, in which the recombinant hGH is administered once two weeks with a dosage of 0.15 to 0.4 mg per weight kg of a patient.
  • the recombimant hGH, GX-H9 may include an amino acid sequence of SEQ ID NO: 1.
  • the pharmaceutical composition of the present disclosure may be administered subcutaneously.
  • yet another aspect of the present disclosure provides a method for treating patients with growth hormone deficiency comprising administering an recombinant hGH GX-H9 to a patient with growth hormone deficiency once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient.
  • Still another aspect of the present disclosure provides a method for treating patients with growth hormone deficiency comprising administering an recombinant hGH GX-H9 to a patient with growth hormone deficiency twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient.
  • hGH-hyFc The recombinant hGH “GX-H9” used in the present application is referred to as hGH-hyFc which is a human growth hormone fused to hybrid Fc and may have an amino acid sequence of SEQ ID NO: 1.
  • the recombinant hGH GX-H9 may be prepared by a method disclosed in US Patent Registration No. 8,529,899.
  • the pharmaceutical composition containing the recombinant hGH GX-H9 may be administered to adults with growth hormone deficiency.
  • the adult growth hormone deficiency means a case where a standard deviation score (SDS) compared to normal people in the same age group in adults is -2 or less ( ⁇ -2 SDS) or within 2.5 percentile ( ⁇ 2.5 percentile).
  • SDS standard deviation score
  • ⁇ -2 SDS ⁇ -2 SDS
  • ⁇ 2.5 percentile 2.5 percentile
  • the adult grown hormone deficiency may be classified into three categories. The adult grown hormone deficiency may be divided into first, childhood-onset growth hormone deficiency, case of grown hormone deficiency due to hypothalamic-pituitary-based disorders, and idiopathic grown hormone deficiency.
  • the pharmaceutical composition of the present disclosure may include a pharmaceutically acceptable carrier that may be any non-toxic material suitable for delivering the recombinant hGH to the patient.
  • a pharmaceutically acceptable carrier such as any non-toxic material suitable for delivering the recombinant hGH to the patient.
  • Distilled water, alcohol, fats, waxes and inert solids may be included as the carrier.
  • Pharmaceutically acceptable adjuvants such as a buffer, a dispersant, and a diluent, for example, bacteriostatic water for injection (BWFI), phosphate buffered saline, a ringer's solution, a dextrose solution, sucrose, poloxamer, and the like may be included in the pharmaceutical composition of the present disclosure.
  • BWFI bacteriostatic water for injection
  • phosphate buffered saline a ringer's solution
  • a dextrose solution sucrose, poloxamer, and the like
  • the recombinant hGH, GX-H9 may be administered once a week with a dosage of 0.1 to 0.3 mg per weight kg of the patient and for example,the administrative dose may vary with dosage of 0.1, 0.15, 0.2, 0.25, or 0.3 mg per weight kg according to age, gender, and estrogen states of patients.
  • the recombinant hGH GX-H9 may be administered once a week with a dosage of 0.1 to 0.2 mg per weight kg of the patient.
  • the recombinant hGH GX-H9 may be administered twice-monthly with a dosage of 0.1 to 0.4 mg per weight kg of the patient and for example, may be administered once two weeks with a dosage of 0.1, 0.15, 0.2, 0.25, 0.3, 0.35, or 0.4 mg per weight kg of the patient according to age, gender, and estrogen states.
  • the recombinant hGH GX-H9 may be administered twice-monthly with a dosage of 0.15 to 0.4 mg per weight kg of the patient.
  • the preferred dosage of the recombinant hGH, GX-H9 is 0.1 mg/kg administered once a week, 0.2 mg/kg administered twice-monthly, or 0.3 mg/kg administered twice-monthly. Further, in some cases, the recombinant hGH may be administered twice-monthly, once in three weeks, or monthly with 0.3 to 0.6 mg/kg according to the age, gender, and estrogen states.
  • the dosage of the recombinant hGH may be adjusted based on the age, gender, and estrogen state of the patient and may be increased or decreased while monitoring the progress of administration.
  • the dosage of the recombinant hGH administered subsequently may be higher or lower than an initial dosage or equal to the initial dosage as level of IGF-1 SDS changes.
  • a small amount of recombinant hGH should be administered safely and then may gradually be increased after verifying no adverse reaction.
  • the dosage of the recombinant hGH may be adjusted while the level of IGF-1 SDS in change is monitored in a plasma or serum sample of the patient.
  • the dosage of the recombinant hGH suitable for each individual patient may vary depending on age, gender, constitution, body weight of the patient.
  • the pharmaceutical composition of the present disclosure containing the recombinant hGH, GX-H9 may be administered to a subject by various methods.
  • the pharmaceutical composition may be administered parenterally and for example, subcutaneously, intramuscularly or intravenously.
  • the composition may be sterilized according to a generally well-known sterilization technique.
  • the composition may include pharmaceutically acceptable auxiliary substances and adjuvants, toxic modifiers and analogs thereof required for adjusting physiological conditions such as pH adjustment, and for example, may include sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, and the like.
  • the concentration of the recombinant hGH may be very various and may be selected preferentially based on a body fluid volume, viscosities, and the like according to the selected specific administration method.
  • the recombinant hGH, GX-H9 may be prepared according to a method disclosed in US Patent Registration No. 8,529,899.
  • a nucleic acid sequence of hGH-hyFc in which hyFc was fused to human growth hormone (hGH) encoding an amino acid sequence of SEQ ID NO: 1 was inserted to an expression vector pAD15 to produce cell line expressing hGH-hyFc.
  • GenBank AAA98618.1 sequence was used for human growth hormone hGH gene and a gene of hyFc was produced by fusing GenBank P01880 (IgD) and GenBank AAH25985 (IgG4) sequences. Genes obtained through gene manufacturer were injected by using specific restriction enzymes as expression vectors for preparation of cell line production.
  • the expression vector ensured by the method above was transfected into CHO DG44 (Columbia University, USA) cells by a calcium phosphate method. After 6 hrs of the transfection, the transfected cells were washed with a phosphate buffer and then a medium was replaced with 10% dFBS (Gibco, USA, 30067-334), MEM alpha (Gibco, 12561, USA, Cat No. 12561-049), and HT+ (Gibco, USA, 11067-030) media. After 48 hrs of the transfection, HT selection was performed by continuously diluting the transfected cells in a 100 mm plate using a 10% dFBS + MEM alpha medium without HT.
  • LDC limiting dilution cloning
  • target protein was purified from the culture solution.
  • a protein culture solution sample went through sample binding using the protein culture solution using Prosep ® Ultra Plus (Merck)and was equilibrated by using 50 mM sodium phosphate, 150 mM sodium chloride and a pH 7.0 buffer.
  • XK16/20 column (GE Healthcare) was used and the protein was eluted by using 100 mM sodium citrate, 200 mM L-arginine and pH 3.1 buffer.
  • Binding force between C1q and GX-H9 inducing cell-mediated cytotoxicity was also measured by using the above ELISA method.
  • As positive control groups Rituxan (by Roche Corporation in Switzerland) and Enbrel (by Amgen Corporation in USA) was used and binding force between the test substances was measured by using the HRP conjugated anti-C1q antibody.
  • the GX-H9 had low binding force with the Fc ⁇ receptor I inducing the antibody-dependent cell-mediated cytotoxicity as illustrated in FIG. 1 and low binding force with C1q inducing the complement-mediated cytotoxicity as illustrated in FIG. 2.
  • GX-H9 Efficacy of GX-H9 was tested by using hypophysectomized rats as an animal disease model.
  • Genotropin Pfizer Corporation, USA
  • GX-H9 was administered once a week and then efficacy was compared.
  • a test was performed by targeting a weight gain of 10% or less for about 1 week after hypophysectomization.
  • Group 1 was a negative control group and subcutaneously administered with only a formulation buffer for two weeks.
  • Group 2 was subcutaneously administered with Genotropin by 0.2 mg/kg every day.
  • Group 3 was administered once a week with Genotropin of 1.4 mg/kg which was a weekly dosage.
  • Group 4 was administered once a week with GX-H9 of 1.4 mg/kg (corresponding to a weekly dosage of Genotropin).
  • Group 5 was administered once a week with GX-H9 of 3.5 mg/kg (corresponding to 1/2 of a molar number of a weekly dosage of Genotropin).
  • Group 6 was administered once a week with GX-H9 of 7.0 mg/kg (corresponding to the same molar number as a weekly dosage of Genotropin). After administration of the drug, symptoms were observed and weights were measured every day.
  • GX-H9 was administered to rats once subcutaneously.
  • Eutropin LG Life Sciences, Inc., in Korea
  • Group 1 was administered with a single dosage of 200 ug/kg of Eutropin subcutaneously
  • Group 2 was administered with a single dosage of 200 ug/kg of GX-H9 subcutaneously
  • Group 3 was administered with a single dosage of 1,000 ug/kg of GX-H9 subcutaneously.
  • Blood was taken before and for 1, 4, 8, 12, 18, 24, 36, 48, 72, 96, 120, 144, 168, 216, 264 and 336 hours after subcutaneous administration.
  • the blood concentration of each substance was measured by specific bioassay (ELISA).
  • test results were illustrated in FIG. 4 and pharmacokinetics after single subcutaneous administration of GX-H9 at a dosage of 200 or 1,000 ug/kg were reached the highest blood concentration at 17 or 24 hours (Tmax) and the GX-H9 was detected in the blood until 9 days and 11 days. As the dose increased, systemic exposure also increased.
  • GX-H9 and Eutropin as a control substance were analyzed in cynomolgus monkey.
  • GX-H9 was subcutaneously administered repeatedly four times a week with dosages of 500 ⁇ g/kg and 1000 ⁇ g/kg and Eutropin as a control substance was subcutaneously administered once at a dosage of 1000 ⁇ g/kg.
  • the blood concentration was measured by specific bioassay (ELISA) for GX-H9 and Eutropin and the results were illustrated in FIGS. 5 and 6.
  • ELISA specific bioassay
  • GX-H9 As compared with Eutropin (1000 ⁇ g/kg, single subcutaneous administration), administeration of GX-H9 (500 or 1000 ⁇ g/kg) was detected (12 to 18 hrs after administration of Eutropin vs 168 hrs after administration of GX-H9) for a longer time in the blood. That is, when GX-H9 is administered subcutaneously, it was observed that the systemic exposure was more persistent than that of control drug, Eutropin. In addition, as the dosage of GX-H9 increased from 500 to 1000 ⁇ g/kg, it was observed that the systemic exposure after subcutaneous administration increased in proportion to the dose increase.
  • Phase 1 clinical study of a randomized, double-blind, placebo-controlled, single ascending dose was performed by targeting healthy volunteers.
  • the purpose of the Phase 1 clinical study was to evaluate safety, tolerability, and phamacodynamic/pharmacokinetic characteristics after single subcutaneous administration of GX-H9.
  • evaluation was performed for a total of 56 days after single subcutaneous administration of GX-H9 to four dosage groups (0.2, 0.4, 0.8 and 1.6 mg/kg).
  • the blood concentration was measured by the specific bioassay (ELISA) of GX-H9 and the results were illustrated in Table 1 below and FIG. 7.
  • a peak at a geometric mean concentration was observed at about 12 hours (8 to 16 hours) after single subcutaneous administration of GX-H9, and a second peak at a lower concentration was observed at about 32 hours (28 to 32 hours) after administration. At the maximum dosage, the second peak corresponded to C max (see FIG. 7). C max and AUC were increased more the dosage over all dosages.
  • a half-life (t1/2) was from 69.2 hrs to 138 hrs and there was a individual variability.
  • FIG. 8 illustrates a change amount of the concentration (ng/mL) of IGF-1 in the blood as compared with a baseline of groups administered with placebo and 0.2, 0.4, 0.8 and 1.6 mg/kg of GX-H9.
  • the concentration of IGF-1 in the blood was increased in proportion to the dosages.
  • the mean maximum increase (% change to baseline) was 81%, 157%, 301% and 349% at the dosage of 0.2, 0.4, 0.8 and 1.6 mg/kg, respectively.
  • the time reaching the highest concentration of IGF-1 in the blood was 48 to 96 hours and was increased in proportion to the dosage. It was verified that the mean concentration of IGF-1 was recovered to the baseline at the 7-th day after administration at the dosage of 0.2 mg/kg and at the 14-th day at other dosages.
  • Table 2 summarized the results of treatment emergent adverse events observed in subjects according to the administered drug, relation between the drug and the adverse events, and the intensity of the adverse events.
  • n Numebr of individuals showing adverse events
  • the dosage was repeatedly administered for a total of 12 weeks with 0.1 mg/kg weekly (Group 1), 0.3 mg/kg twice-monthly (Group 2), or 0.2 mg/kg twice-monthly (Group 3).
  • Genotropin was administered daily by 6 ⁇ g/kg (G: Group 4).
  • the blood was taken during the first administration (1st week) and the last administration (12th week) and taken at 1, 2, 4, 8, 12, 18, 24, 48, 72 and 168 hrs after administration.
  • the blood was taken before the first administration (first week) and the last administration (11-th week) and taken at 1, 2, 4, 8, 12, 18, 24, 48, 72, 168, 240 and 336 hrs after administration.
  • the concentration of the GX-H9 in the blood was measured from the obtained blood samples and the results were illustrated in Tables 4 to 6 and FIG. 9.
  • the change in pharmacokinetics was analyzed at the first administration (1st week) and the last administration (11th week), before administration, and 1, 2, 4, 8, 12, 18, 24, 48, 72, 168, 240 and 336 hrs after administration.
  • IGF-1 SDS IGF-1 standard deviation score
  • the blood was taken during the first administration (1st week) and the last administration (12th weeks) and taken at 12, 24, 48, 72 and 168 hrs after administration.
  • IGF-1 SDS IGF-1 standard deviation score
  • the IGF-1 SDS was anlayzed at 4 days after administration of 3, 5, 7, 9, and 11-th weeks.
  • the group administered with GX-H9 twice-monthly the blood was taken during the first administration (1st week) and the last administration (11th weeks) and taken at 12, 24, 48, 72, 168, 240 and 336 hrs after administration.
  • the change of IGF-1 SDS was analyzed at 4 days after administration of 3, 5, 7, and 9-th weeks.
  • the change in pharmacodynamics was analyzed at the first administration (1st week) and the last administration (11th week), before administration, and 12, 24, 48, 72, 168, 240 and 336 hrs after administration.
  • the mean (standard deviation)IGF-1 SDS of -2.64 (1.25) and -2.40 (0.82) before administration showed change with minimum of -1.36 (1.59) and -2.01 (0.88)and maximum of 2.12 (1.82) and 1.33 (1.83) within 14 days after the last administration (11th week).
  • the maximum mean change in IGF-1 SDS of GX-H9 was shown between 48 hours to 72 hours after administration of 0.1 mg/kg at weekly interval or 0.3 mg/kg at a twice-monthly interval and the minimum mean change was shown within 168 to 336 hrs after administration. Meanwhile, in Genotropin, an active control drug, which was administered daily, the maximum mean change was shown within 12 hrs after administration and the minimum mean change was reached within 24 hrs after administration.
  • the goal of treatment for adult patients with growth hormone deficiency is to improve metabolism normalization and quality of life.
  • the IGF-1 level in the blood after administration of GX-H9 needs to be optimized to the middle (50th percentile or 0 SDS) to 1 SDS range within the normal range (-2 SDS to 2 SDS). Accordingly, in the case of administering GX-H9 once a week, it was verified that the range of IGF-1 SDS of the treated patient may be maintained in the normal level by adjusting the treatment dosage in a concentration range of 0.1 mg/kg to 0.2 mg/kg according to IGF-1 level.
  • the range of IGF-1 SDS of the treated patient may be maintained in the normal level by adjusting the treatment dosage in a concentration range of 0.2 mg/kg to 0.4 mg/kg according to IGF-1 level.
  • n Numebr of individuals showing adverse events
  • % inhibition is defined as 100 x (1-(mean OD GX-H9 spiked/mean OD unspiked sample)); if ⁇ 17% inhibition sample will be assessed for titer3)
  • Titer value titer sample is defined as the reciprocal of the dilution that generates a mean OD greater than or equal to the cutpoint OD of the plate where the subsequent dilutions in the series results in a mean OD less than the cutpoint OD
  • the GX-H9 showed the equivalent efficacy as the growth hormone in the body or the first-generation growth hormone product and had an enhanced half-life, and thus convenience of medication was very improved and safety was verified.

Abstract

La présente invention concerne un procédé d'administration d'une hormone de croissance humaine recombinante GX-H9 pour traiter un déficit en hormone de croissance. En particulier, la présente invention concerne une composition pharmaceutique pour traiter un déficit en hormone de croissance, contenant un hGH recombinant GX-H9 et un véhicule pharmaceutiquement acceptable, dans laquelle le GX-H9 recombinant est administré une fois par semaine avec une dose de 0,1 à 0,3 mg par kg de poids d'un patient ou deux fois par mois avec une dose de 0,1 à 0,4 mg par kg de poids du patient. En outre, la présente invention concerne un procédé de traitement d'un déficit en hormone de croissance comprenant l'administration d'un hGH recombinant GX-H9 à un patient atteint d'un déficit en hormone de croissance une fois par semaine avec une dose de 0,1 à 0,3 mg par kg de poids du patient ou deux fois par mois avec une dose de 0,1 à 0,4 mg par kg de poids du patient.
PCT/KR2017/001726 2016-02-17 2017-02-16 Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance WO2017142331A1 (fr)

Priority Applications (10)

Application Number Priority Date Filing Date Title
JP2018543675A JP6896749B2 (ja) 2016-02-17 2017-02-16 組換えhGHを含む、成長ホルモン欠乏を治療するための薬学組成物
RU2018132694A RU2732113C2 (ru) 2016-02-17 2017-02-16 Фармацевтическая композиция, включающая рекомбинантный гормон роста человека для лечения дефицита гормона роста
CA3014164A CA3014164A1 (fr) 2016-02-17 2017-02-16 Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un deficit en hormone de croissance
PL17753492.2T PL3416677T3 (pl) 2016-02-17 2017-02-16 Kompozycja farmaceutyczna obejmująca rekombinowany hgh do leczenia niedoboru hormonu wzrostu
DK17753492.2T DK3416677T3 (da) 2016-02-17 2017-02-16 Farmaceutisk sammensætning omfattende rekombinant hgh til behandlingen af væksthormonmangel
CN201780018326.9A CN109152816B (zh) 2016-02-17 2017-02-16 用于治疗生长激素缺乏症的包含重组hgh的药物组合物
BR112018016817A BR112018016817A2 (pt) 2016-02-17 2017-02-16 composição farmacêutica e método de tratamento da deficiência do hormônio do crescimento
ES17753492T ES2925901T3 (es) 2016-02-17 2017-02-16 Composición farmacéutica que comprende hGH recombinante para el tratamiento de la deficiencia de hormona del crecimiento
US16/077,177 US20210177945A1 (en) 2016-02-17 2017-02-16 Pharmaceutical composition comprising recombinant hgh for the treatment of growth hormone deficiency
EP17753492.2A EP3416677B1 (fr) 2016-02-17 2017-02-16 Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR20160018695 2016-02-17
KR10-2016-0018695 2016-02-17
KR1020170021104A KR20170096968A (ko) 2016-02-17 2017-02-16 hGH 융합단백질을 포함하는 성장호르몬 결핍을 치료하기 위한 약학 조성물
KR10-2017-0021104 2017-02-16

Publications (1)

Publication Number Publication Date
WO2017142331A1 true WO2017142331A1 (fr) 2017-08-24

Family

ID=59626140

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/001726 WO2017142331A1 (fr) 2016-02-17 2017-02-16 Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance

Country Status (1)

Country Link
WO (1) WO2017142331A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018044060A1 (fr) * 2016-08-30 2018-03-08 Genexine,Inc. Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh
EP3506923A4 (fr) * 2016-08-30 2020-04-22 Genexine, Inc. Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7867491B2 (en) 2007-05-30 2011-01-11 Genexine Co., Ltd. Immunoglobulin fusion proteins
US20140162949A1 (en) * 2012-06-05 2014-06-12 Amunix Operating Inc. Treatment with human growth hormone analogues
US8883134B2 (en) * 2010-10-20 2014-11-11 Handok Pharmaceuticals, Inc. Human interleukin-1 receptor antagonist—hybrid Fc fusion protein

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7867491B2 (en) 2007-05-30 2011-01-11 Genexine Co., Ltd. Immunoglobulin fusion proteins
US8529899B2 (en) 2007-05-30 2013-09-10 Genexine, Inc. Human growth hormone immunoglobulin fusion proteins
US8883134B2 (en) * 2010-10-20 2014-11-11 Handok Pharmaceuticals, Inc. Human interleukin-1 receptor antagonist—hybrid Fc fusion protein
US20140162949A1 (en) * 2012-06-05 2014-06-12 Amunix Operating Inc. Treatment with human growth hormone analogues

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
"Consensus guideline for the diagnosis and treatment of GH deficiency in childhood and adolescence: summary statement of the GH Research Society. GH Research Society", J. CLIN. ENDOCRINOL. METAB., vol. 85, no. 11, November 2000 (2000-11-01), pages 3990 - 3
"Guidelines for Use of Growth Hormone in Clinical Practice", ENDOCR. PRACT., vol. 15, no. 2, 2009
ENDOCRINE PRACTICE, vol. 14, no. 2, March 2008 (2008-03-01), pages 143 - 54
HOYBYE ET AL.: "Status of Long-Acting-Gowth Hormone Preparations - 2015", GROWTH HORMONE & IGF RESEARCH, vol. 25, 14 July 2015 (2015-07-14), pages 201 - 206, XP055273438 *
KIM ET AL.: "Controlled Release of Human Growth Hormone Fused with a Human Hybrid Fc Fragment Through a Nanoporous Polymer Membrane", NANOSCALE, vol. 5, 2013, pages 4262 - 4269, XP055411456 *
KIM ET AL.: "Pharmacokinetics, Pharmacodynamics, and Efficacy of a Novel Long-Acting Human Growth Hormone:Fc Fusion Protein", MOLECULAR PHARMACEUTICS, vol. 12, 15 September 2015 (2015-09-15), pages 3759 - 3765, XP055575158 *
PLOS ONE, vol. 6, no. 1, January 2011 (2011-01-01), pages e16223

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018044060A1 (fr) * 2016-08-30 2018-03-08 Genexine,Inc. Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh
EP3506923A4 (fr) * 2016-08-30 2020-04-22 Genexine, Inc. Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh
TWI718334B (zh) * 2016-08-30 2021-02-11 南韓商格納西尼有限公司 一種人類生長激素(hgh)融合蛋白gx-h9作為藥物的用途及試劑盒

Similar Documents

Publication Publication Date Title
AU2016346864B2 (en) Long-acting FGF21 fusion proteins and pharmaceutical composition comprising same
AU2015268199B2 (en) Composition for treating diabetes mellitus comprising insulin and a GLP-1/glucagon dual agonist
WO2012165915A2 (fr) Composition pour le traitement du diabète comprenant un conjugué d'insuline à action prolongée et un conjugué de peptide insulinotropique à action prolongée
WO2016108654A1 (fr) Polypeptide de fusion de glp et d'un fc hybride d'immunoglobuline, et utilisation de ce dernier
WO2014017847A1 (fr) Formulation liquide à base d'un conjugué d'insuline à action prolongée
WO2014017849A1 (fr) Formulation liquide à base d'insuline à action prolongée et d'un peptide insulinotrope
EP2552474A2 (fr) Conjugué d'insuline utilisant un fragment d'immunoglobuline
WO2016114633A1 (fr) Protéines de fusion fgf21 à action prolongée et composition pharmaceutique les comprenant
WO2016108586A1 (fr) Dérivé du glucagon à stabilité améliorée
WO2018088838A1 (fr) Composition pharmaceutique comprenant des protéines de fusion destinée à la prévention ou le traitement de l'hépatite, de la fibrose hépatique et de la cirrhose hépatique
WO2017142331A1 (fr) Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance
AU2015372767B2 (en) Glucagon derivatives
WO2018044060A1 (fr) Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh
EP3506923A1 (fr) Composition pharmaceutique destinée à traiter une déficience en hormone de croissance contenant une protéine de fusion de hgh
EP3416677A1 (fr) Composition pharmaceutique comprenant hgh recombinant pour le traitement d'un déficit en hormone de croissance
WO2020116810A1 (fr) Composition pharmaceutique, comprenant un peptide inhibiteur contre la signalisation fas, destinée à la prévention ou au traitement de l'obésité, du foie gras, ou de la stéatohépatite
WO2021107603A2 (fr) Protéine de fusion gdf15 à action prolongée et composition pharmaceutique la comprenant
WO2021162460A1 (fr) Nouvelle composition pharmaceutique pour le traitement d'une maladie hépatique non alcoolique
WO2022245183A1 (fr) Composition pour la prévention ou le traitement d'une stéatose hépatique non alcoolique ou d'une stéatohépatite non alcoolique comprenant un variant de facteur de différenciation de croissance 15
WO2021149945A1 (fr) Protéine de fusion comprenant une protéine pd-l1 et utilisation associée
WO2023059099A1 (fr) Composition pharmaceutique pour la prévention, l'atténuation, le soulagement ou le traitement de la fibrose pulmonaire, comprenant de l'ézétimibe en tant que principe actif
US20240131114A1 (en) Nerve growth factor fusion protein, preparation method and use thereof

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: 17753492

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 3014164

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2018543675

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112018016817

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 2017753492

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017753492

Country of ref document: EP

Effective date: 20180917

ENP Entry into the national phase

Ref document number: 112018016817

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20180816