Movatterモバイル変換


[0]ホーム

URL:


WO2025056188A1 - Relaxin medication - Google Patents

Relaxin medication
Download PDF

Info

Publication number
WO2025056188A1
WO2025056188A1PCT/EP2023/082763EP2023082763WWO2025056188A1WO 2025056188 A1WO2025056188 A1WO 2025056188A1EP 2023082763 WEP2023082763 WEP 2023082763WWO 2025056188 A1WO2025056188 A1WO 2025056188A1
Authority
WO
WIPO (PCT)
Prior art keywords
relaxin
medicament
intended
treatment
cells
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
PCT/EP2023/082763
Other languages
French (fr)
Inventor
Thomas Bernd DSCHIETZIG
Ulrich M.E. BENEDUM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Relaxera Pharmazeutische GmbH and Co KG
Original Assignee
Relaxera Pharmazeutische GmbH and Co KG
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 Relaxera Pharmazeutische GmbH and Co KGfiledCriticalRelaxera Pharmazeutische GmbH and Co KG
Publication of WO2025056188A1publicationCriticalpatent/WO2025056188A1/en
Pendinglegal-statusCriticalCurrent
Anticipated expirationlegal-statusCritical

Links

Classifications

Definitions

Landscapes

Abstract

Relaxera Pharmazeutische GmbH & Co.KG 36 IDK00257PC 20-Nov-23 SUMMARY. A pharmaceutical composition for daily (q.d.) or twice daily (b.i.d.) subcutaneous injection of a bolus of human relaxin-2 or a complex of relaxin-2 and zinc for a period of at least one week, preferably three to twelve months, and injection pens for treating heart failure with preserved ejection fraction, atrial fibrillation, a patient suffering from diabetes and heart failure with preserved ejection fraction, and/or atrial fibrillation; for producing the protective effects of synthetic human relaxin-2 associated with ischemic heart disease, including the reduction of inflammatory leukocyte and platelet responses, inhibition of the release of proinflammatory and arrhythmogenic mediators by leukocytes and mast cells and other harmful substances generated by inflammation, oxidative stress, and necrosis, for use as a broad-spectrum replacement of glucocorticoids, mineralocorticoids, corticosteroids and their analogues in the treatment of tissue and endothelial injury without inducing gluconeogenesis and cushingoid adverse effects; for supporting the repair of tissue and endothelial injuries by promoting immunosuppressive regulatory T cells (Treg), particularly, in autoimmune pathologies and organ transplantation. Figure 4D

Description

RELAXIN MEDICATION
FIELD OF THE INVENTION
[001] The present invention relates to a medication containing a relaxin peptide of the human insulin superfamily and a dosing regimen for treating patients suffering from heart failure (HF), notably heart failure with preserved ejection fraction (HFpEF), or a condition requiring a dampening of the innate immune system (A61 K 38/1754; A61 K 38/1751 ; A61 K 38/2221).
BACKGROUND OF THE INVENTION
[002] Heart failure (HF) is commonly classified according to which heart function is most affected. Left-sided heart failure describes a failure of the left-sided pumping action which moves oxygen-rich blood from the lungs through the left atrium into the left ventricle and then out to the rest of the body. The term right-sided heart failure is used to describe a failure of the right-sided pumping action which pumps blood returning to the heart through the veins, through the right atrium into the right ventricle, and then back out to the lungs to be replenished with oxygen. Symptoms of heart failure are further distinguished by whether they developed quickly (acute heart failure) or gradually over time (chronic heart failure). Congestive heart failure (CHF) refers to a condition in which the heart cannot pump enough blood to meet the body's needs. As blood flow out of the heart slows, blood returning to the heart through the veins backs up, causing congestion in the tissues. Typical signs of CHF include swollen legs or ankles, fatigue, pulmonary edema, respiratory distress. CHF also affects the kidney function and can be caused by coronary artery disease, congenital and primary heart disease, infections of the heart muscle (cardiomyopathy, endocarditis and/or myocarditis), myocardial infarction, high blood pressure, heart valve disease. The drugs used to treat CHF are angiotensin inhibitors, vasodilators, and mineralocorticoid antagonists (e.g. spironolactone), SGLT-2 inhibitors (e.g. empagliflozin) to dilate blood vessels and reduce resistance, beta blockers to improve the function of the left ventricle, digitalis to increase the pumping action of the heart and diuretics to eliminate excess salt and water.
[003] The present application concerns heart failure with preserved ejection fraction (HFpEF), which is also referred to as heart failure with normal ejection fraction or diastolic heart failure (DHF) because this subgroup of heart failure is associated with a decline in diastolic performance of the left ventricle of the heart. HFpEF is characterized histologically by cardiomyocyte hypertrophy, increased interstitial collagen deposition and calcium deposition within the myocardium which together are thought to result in reduced distensibility and compliance.
[004] Heart Failure (HF) is the leading cause of cardiovascular death and ~50% of
HF patients suffer from HFpEF. Patients with chronic HFpEF have a near-normal ejection fraction, reduced left ventricular (LV) diastolic compliance, elevated LV pressure, fibrosis, and ion channel remodeling leading to supraventricular arrhythmias that increase morbidity and mortality. HFpEF patients typically have comorbidities such as atrial fibrillation (AF), diabetes, pulmonary edema, and hypertension. The latter is a predictor of mortality and is targeted to prolong survival in the absence of a direct therapy for HFpEF. Exogenous human relaxin (RLX, RLN2) which addresses most aspects of clinical HFpEF has been proposed as a solution as it act as an endocrine and paracrine factor that dilates blood vessels and increases blood flow in tissues (see Dschietzig T et al. in Relaxin: a pregnancy hormone as a central player of body fluid and circulation homeostasis, CMLS 2003; 60:688-700; Dschietzig T et al. in Relaxin - a pleiotropic hormone and its emerging role for experimental and clinical therapeutics, Pharmacol Ther 2006; 112:38e56; US 10,226,512B2; US 2015/0232526A1 ,
US 2017/0342120A1 , US 2020/0087371 A1 , WO 2014/059174A1 ; US 7,878, 978B2,
US 8,602,998B2; EP 1 253 929B1 ; US 6,723,702B2; WO 2001/058468A1 ; EP 3 347 037B1 ;
WO 2017/042577A1 ; WO 2022/037469A1 ; Muppidi, A. et al, Design and Synthesis of Potent,
Long-Acting Lipidated Relaxin-2 Analogs, BIOCONJUGATE CHEM., Vol. 30 (2018-12-13), pp
83-89; Nair, V.B. et al., Synthetic Covalently Linked Dimeric Form ofH2 Relaxin Retains Native
RXFPI Activity and has improved in Vitro Serum Stability", BiOMED RESEARCH INT. (2015-
12-31); US 2014/0057832 A1 ; US 2002/0019349 A1). No effective galenical form of relaxin-2 is available (see Bani D in Recombinant human H2 relaxin (serelaxin) as a cardiovascular drug: aiming at the right target, Drug Discovery Today (2020) 25(7), 1239-44;
EMA/303748/2014 - Assessment report, EMA/299385/2014). [005] Relaxin, originally described as a pregnancy hormone, is a heterodimeric peptide of approx. 6 kDa and a member of the insulin superfamily which includes insulin, insulin-like growth factors I and II, relaxin-1 , -2, and -3; and insulin-like factors 3, 4, 5, and 6.
This superfamily has no common primary sequence homology, but relaxin has intra- and interchain disulfide bridges linking an A chain and a B chain, as in insulin. In humans, three different forms of relaxin have been identified that do not share similar peptide sequences.
Relaxin-2 is the only form that is secreted into the circulation. [006] Besides being a maternal-fetal interface hormone (Hisaw FL in Experimental relaxation of the pubic ligament of the guinea pig, Proc. Soc. Exp. Biol. Med. 1926; 23:661- 663), numerous clinical applications of relaxin have been proposed: for the treatment of cutaneous aging, androgenetic alopecia, atrophy, sclerosis, and miniaturization of the hair and hair follicles (EP0793505); for control of fetal growth (EP0991947), for increasing fertility
(EP1473034), as an adjuvant in the differentiation of stem cells (EP1696948), for increasing arterial compliance (EP1765149), for diseases related to vasoconstriction (EP1854476), for tumor suppression (W02007115414), for the treatment of diabetes and related complications (EP1909809), for treating multiple sclerosis and other neurodegenerative dysfunctions
(EP2723366), for treating symptoms of aging and neurodegenerative dysfunctions (W00048618), for treating glucotoxicity and impaired glucose tolerance (EP2817026), in treating dyspnea associated with acute heart failure (EP2829280), for treating heart failure with preserved ejection fraction (EP3145534), for treating CNS, CNS trauma, demyelinating disease and/or gliosis, multiple sclerosis (MS), Alzheimer’s disease and Parkinson’s disease, inflammatory conditions of CNS, diffuse cerebral sclerosis of Schilder; acute disseminated encephalomyelitis, acute hemorrhagic leukoencephalitis, transverse myelitis, and neuromyelitis optica, concussion, traumatic brain injury, shaken baby syndrome, traumatic spinal cord injury, traumatic brain injury, ionizing radiation, Korsakoff’s syndrome, multiple systemic atrophy, prion disease, AIDS dementia complex, vasculitis, amyotrophic lateral sclerosis, Huntington’s disease, autoimmune inflammatory disorders, retinal gliosis, encephalopathies, leukodystrophies, encephalitis, neuropathies (EP3347037), for treating various inflammatory conditions, acidic airway hyperreactivity, asthma, rheumatoid arthritis, gout, ankylosing spondylitis, inflammatory enteritis, myositis, systemic lupus erythematosus, sepsis, urticaria, psoriasis, allergic reactions (WO20220374669). A particular focus has been the use of relaxin for hemodynamic adaption and regulation of systemic vascular resistance in models of cardiac, renal, pulmonary, and hepatic infarction (WO9303755, W00240500;
Dschietzig T et al. in Plasma levels and myocardial expression of relaxin-2 are increased in human heart failure, CIRCULATION 2000, 102(18):594; Coulson CC et al. in Central hemodynamic effects of recombinant human relaxin in the isolated, perfused rat heart model,
Obstetrics & Gynecology 1996, 87(4):610-612; Masini E. et al. in Relaxin counteracts myocardial damage induced by ischemia-reperfusion in isolated guinea pig hearts: evidence for an involvement of nitric oxide, Endocrinology 1997, 138:4713-4720; DiLascio G et al. in Cellular retrograde cardiomyoplasty and relaxin therapy for postischemic myocardial repair in a rat model, Tex Heart Inst J 2012, 39:488-499; Collino M et al. in Acute treatment with relaxin protects the kidney against ischemia-reperfusion injury., J Cell Mol Med 2013, 17:1494-1505;
Bausys A et al. in Custodiol® supplemented with synthetic human relaxin decreases ischemia- reperfusion injury after porcine kidney transplantation, Int J Mol Sci. 2021 , 22, 11417; Alexiou K et al. in Relaxin is a candidate drug for lung preservation: relaxin induced protection of rat lungs from ischemia-reperfusion injury, J Heart Lung Transplant 2010, 29:454-460; Teichmann SL et al. in Relaxin: a review of the biology and potential role in treating heart failure, Curr Heart
Fail Rep 2010; 7:75-82). In addition, relaxin has been observed to reduce oxidative cell damages that occur in orthotopic kidney and liver transplants and liver perfusion systems
(DE102005040492; Boehnert MU in Relaxin as an additional protective model of isolate perfused rat liver, Ann N Y Acad Sci 2005, 1041 :434-440; Kageyama S et al. in Relaxin in Liver
Transplantation: A Personal Perspective, Mol Cell Endocrinol. 2019, 487: 75-79;
Jakubauskiene L et al. in Relaxin positively influences ischemia-reperfusion injury in solid organ transplantation: a comprehensive review, Int J Mol Sci. 2020, 21 (2):631ff). These findings appear to be consistent with observations that relaxin can act as a ligand of the GR in somatic cells, completely independent of the signaling cascades of the G-protein-coupled relaxin receptors RXFP1 and RXFP2 (formerly designated LGR7 and LGR8, respectively).
Experiments in Hela cells, HEK cells and Th1-activated macrophages show that relaxin-2 can activate the GR and that the active relaxin-GR complex depresses stimulated secretion of the cytokines IL-1 , IL-6, and TNF-a like dexamethasone (Dschietzig TB et al. in Identification of the pregnancy hormone relaxin as a glucocorticoid receptor agonist, FASEB J 2004, 18:1536- 1538; Dschietzig T et al. in The pregnancy hormone relaxin binds to and activates the human glucocorticoid receptor, Ann N Y Acad Sci. 2005, 1041 :256-71 ; Dschietzig T et al. in RXFP1- inactive relaxin activates human glucocorticoid receptor: further investigations into the relaxin-
GR pathway, Regul Pept. 2009, 154:77-84; Dschietzig T et al. in Autoregulation of human relaxin-2 gene expression critically involves relaxin and glucocorticoid receptor binding to glucocorticoid response half-sites in the relaxin-2 promoter, Regul Pept. 2009, 155:163-73).
Glucocorticoids and corticosteroids are effective immunosuppressants in treating inflammatory, autoimmune, and lymphoproliferative diseases. At all cellular level, they are functionally mediated by the glucocorticoid receptor (GR). The functions of the activated GR complexes are pleiotropic and occur in various parts of the body: in the control of metabolism, body development, and immune response. This has led to steroids and glucocorticoids being the most used drugs (see for review Nicolaides N et al., Glucocorticoid Receptor in Feingold
KR, Anawalt B, Boyce A, et al., eds. Endotext [Internet], South Dartmouth (MA): 2021). Because almost all cells in the body express the same glucocorticoid receptor, the pharmacologically highly desirable effects of glucocorticoids and GR activation are equally associated with specific adverse effects caused thereby. Adverse metabolic and pharmacologic effects of prolonged glucocorticoid treatment include wound healing disorders, manifestation/deregulation of diabetes mellitus, adverse immunosuppression, increased risk of infection, osteoporosis, growth disturbances in children as well as myopathy/muscular atrophy, skin atrophy, steroid acne, hirsutism in addition to the typical symptoms of a Cushing’s syndrome such as uncontrollable hypertension, disturbances of salt and water balance, psychological and neurological disorders, depression, etc. Therefore, an individual benefit-risk analysis must be performed for each glucocorticoid therapy. This is particularly true for patients who have a chronic inflammatory disease but for whom the harmful side effects of prolonged glucocorticoid treatment would outweigh the painful or debilitating effects of the underlying disease.
[007] The known prior art does not provide useful guidance whether and when treatment with relaxin-2 is medically indicated or a dosage regimen to produce the physiologically hypothesized effects of relaxin-2. There is also no knowledge of an effective pharmaceutical formulation and dosage regimen of relaxin. The prior art represents a problem.
SUMMARY OF THE INVENTION
[008] The problem is solved by a pharmaceutical formulation for daily (q.d.) or twice daily (b.i.d.) subcutaneous injection over a prolonged period of at least one or two weeks comprising as active ingredient dissolved synthetic human relaxin-2 from 75 pg to 2250 micrograms and a pharmacologically acceptable adjuvant, excipient, or diluent.
[009] In some embodiments, the active ingredient for q.d or b.i.d subcutaneous injection over a prolonge8d period is dissolved in 0.5 to 1 .5 ml aqua bidest. as diluent.
[0010] In some embodiments, the active ingredient for q.d or b.i.d subcutaneous injection over a prolonged period is dissolved in 0.5 to 1 .5 ml aqueous sodium acetate pH 5.0 as diluent.
[0011] In some embodiments, the active ingredient for q.d or b.i.d. subcutaneous injection over a period of a prolonged period is dosed as a complex of zinc and human relaxin-
2.
[0012] In some embodiments, the active ingredient for q.d or b.i.d. subcutaneous ejection comprises a hexamer complex of Zn2+ ions and human relaxin-2.
[0013] In some embodiments, the pharmaceutical formulation for daily (q.d.) or twice daily (b.i.d.) subcutaneous injection as described above is for a prolonged period of at least one week, preferably three to twelve months.
[0014] In some embodiments, the pharmaceutical formulation is contained in a pen with a ready-to-use medication pen for subcutaneous injection. Such a pen for subcutaneous injections has a smaller needle to ensure that the medication enters the fatty tissue and not the muscle.
[0015] In some embodiments, the medication for daily (q.d.) or twice daily (b.i.d) subcutaneous injection is for treating heart failure with preserved ejection fraction for a period of at least three months. [0016] In some embodiments, the medication for daily (q.d.) or twice daily (b.i.d) subcutaneous injection is for treating a patient suffering from heart failure with preserved ejection fraction for a period of six months or more.
[0017] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for the treatment of a diabetes patient suffering from heart failure with preserved injection fraction.
[0018] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for the treatment of a diabetes patient suffering from atrial fibrillation and heart failure with preserved injection fraction.
[0019] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for the treatment of ischemic heart disease.
[0020] In some embodiments, the relaxin medication for daily (q.d.) or twice daily (b.i.d) subcutaneous injection is for the treatment of ischemic heart disease.
[0021] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for an adjunctive medical therapy to coronary angioplasty.
[0022] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection of synthetic human relaxin-2 is for the protective actions of relaxin associated with ischemic heart disease, including the reduction of inflammatory leukocyte and platelet responses inhibition of the release of proinflammatory and arrhythmogenic mediators by leukocytes and mast cells and other harmful substances generated locally at reperfusion by inflammation, oxidative stress, and necrosis.
[0023] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for the prolonged treatment of a patient requiring a dampening of the innate immune system, in particular a patient having received a heart or kidney allograft.
[0024] In some embodiments, the relaxin medication for daily (q.d.) or twice daily (b.i.d) subcutaneous injection is for the prolonged dampening of the innate immune system, in particular of inflammatory responses triggered by the innate immune system and/or suppression through a ligand-activated glucocorticoid receptor, whereby manifestation or deregulation of diabetes or the symptoms of a Cushing’s syndrome are avoided.
[0025] In some embodiments, the patient suspected of having inflammatory responses triggered by the innate immune system has the following clinical criteria: pre- diabetes (HbA1C > 5.7 and < 6.5 %), obesity (BMI > 30 kg/m2), hypertension (stage 1 or higher according to the 2017 ACC/AHA Guidelines). [0026] In some embodiments, the injectable relaxin medication is for the prolonged treatment of a patient in need of an altered body or organ development while preventing manifestation or deregulation of diabetes or symptoms of a Cushing’s syndrome. Said patient may display the following clinical characteristics: pre-diabetes (HbA1C > 5.7 and < 6.5 %), obesity (BMI > 30 kg/m2), hypertension (stage 1 or higher).
[0027] In some embodiments, the relaxin medication for daily (q.d.) or twice daily
(b.i.d) subcutaneous injection is for treating a patient having received an allotransplant and in need of a dampening of the innate immune system and inflammatory responses while preventing compromised wound healing, manifestation, or deregulation of diabetes or symptoms of a Cushing’s syndrome. Said medical need of a dampening of the innate immune system is given in case of a transplant patient when one or more of the following four criteria are fulfilled: serum HMGB1 (high-mobility group box protein) greater or equal to 2 ng/ml, serum sTLR4 (soluble Toll-like receptor-4) greater or equal to 0.25 ng/ml, serum sRAGE (soluble receptor of advanced glycation end-products) greater or equal to 0.5 ng/ml, and/or serum calprotectin greater or equal to 4 micrograms/ml.
[0028] In some preferred embodiments, the daily (q.d.) or twice daily (b.i.d) subcutaneous injection of relaxin-2 is for treating a patient in need of chronic suppression of the innate immune system and inflammatory responses while preventing manifestation or deregulation of diabetes or symptoms of a Cushing’s syndrome.
[0029] In some embodiments, said patient suspected of requiring a chronic suppression of inflammatory reactions manifests one or more medical criteria selected from serum HMGB1 (high-mobility group box protein) greater than or equal to 4 ng/ml, serum sTLR4
(soluble Toll-like receptor-4) greater than or equal to 0.5 ng/ml, serum sRAGE (soluble receptor of advanced glycation end-products) greater than or equal to 2 ng/ml, and/or serum calprotectin greater than or equal to 10 micrograms/ml.
[0030] In some other embodiments, the pharmaceutical formulation for daily (q.d.) or twice daily (b.i.d) subcutaneous injection of relaxin-2 is for treating a patient in need of hormone-refractory cancer therapy, including, but not limited to prostate cancer, breast cancer, or a primary cancer therapy through a ligand-activated GR, including, but not limited to Multiple
Myeloma, Hodgkin’s Disease, and other Lymphoid Cancers; Kaposi Sarcoma, the synthetic human relaxin-2 being used as supplement and substitute of the glucocorticoid receptor activating hormone. [0031] Another aspect of the invention relates to a method of treating a patient which comprises testing said patient for one or more of the following clinical parameters: serum
HMGB1 (high-mobility group box protein) greater than or equal to 4 ng/ml; serum sTLR4 (soluble Toll-like receptor-4) greater than or equal to 0.5 ng/ml; serum sRAGE (soluble receptor of advanced glycation end-products) greater than or equal to 2 ng/ml, and/or serum up m calprotectin greater than or equal to 10 micrograms/ml, and when given, administering to the patent an effective amount of synthetic human relaxin-2 within a pharmacological solvent, diluent, or excipient to dampen or suppress physiologic inflammatory reactions by the innate immune system. [0032] Method of treating a patient as described above, who is suffering from forms of autoimmune or rheumatic diseases; ankylosing spondylitis (AS) and spondylarthritis, fibromyalgia, gout, infectious arthritis, lupus, systemic autoimmune disease, osteoarthritis (OA), psoriatic arthritis (PsA) and inflammatory types of arthritis, rheumatoid arthritis (RA).
[0033] Further aspects and advantages of embodiments of the present invention will become apparent from the following detailed description, taken in conjunction with the accompanying drawings, illustrated by way of example of the principles of the invention.
BRIEF DESCRIPTION OF THE DRAWINGS
[0034] In the figures and drawings appended hereto: -
Fig. 1A,B shows a plot (A) of the serum relaxin-2 concentration after a single subcutaneous injection of 30 micrograms/kg (squares), 100 micrograms/kg
(circles) and 500 micrograms/kg (triangles) of synthetic human relaxin-2 into healthy Sprague-Dawley rats over 24 hours, and a plot (B) in which the first four hours have been extended.
Fig. 2A is a sequence and primary structure comparison of the A- and B-chain of human relaxin-2 (SEQ ID NO: 1 and SEQ ID NO: 02) - the side figure B shows the calculated conformation of H2 relaxin - and the sequences of the A- and B- chain of H3 Relaxin (SEQ ID NO:03 and SEQ ID NO:04), the A- and B-chain of insulin-like protein INSL3 (SEQ ID NO:05 and SEQ ID NO:06), insulin-like protein INSL5 (SEQ ID NO: 07 and SEQ ID NO: 08) and human insulin (SEQ
ID NO:09 and SEQ ID NO: 10).
Fig. 2B a plot showing the serum relaxin-2 concentration after a single subcutaneous injection of 1000 micrograms/kg bw of synthetic human relaxin-2 (blue: no complex) and of different Rlx/Zn-complexes with increasing amounts of Zn (orange: Rlx/Zn molar ratio 1 :1 ; grey: 1 :2; yellow: molar ratio 1 :4) in healthy Sprague-Dawley rats; the relaxin bolus of relaxin-Zn complex is considerably longer. Fig. 3A is a bar graph showing the development of maximum velocity of diastolic left ventricular myocardial relaxation E’ in m/s in the ZSF1 rat model of experimental HFpEF as assessed by echocardiography from 10 weeks to 18 weeks (HFpEF)
Fig. 3B is a bar graph showing the ratio E/E’ in the ZSF1 rat model of experimental
HFpEF from 10 weeks to 18 weeks (HFpEF). Fig. 3C is a bar graph comparing the measurable left atrial lengths as a measure of atrial remodeling (PSLAX - parasternal long axis, a specific echo view) of said
ZSF1 rats of experimental HFpEF (10 to 18 weeks) from 10 to 18 weeks.
Fig. 3D is a bar graph graph with the pulmonary oedema score (lung score) derived from lung ultrasound according to Villalba-Orero M et al (Lung ultrasound as a translational approach for non-invasive assessment of heart failure with reduced or preserved ejection fraction in mice, Cardiovascular Research 2017
113:1113-1123) as a measure of the risk of pulmonary oedema in the ZSF1 rat model of experimental HFpEF from 10 to 18 weeks.
Fig. 4A is a bar graph showing the effect on E/E’ in ZSF1 rats with experimental HFpEF
(n = 5-6 per group) of a continuous subcutaneous infusion of 400 pg per kg rat body weight of synthetic human relaxin-2 over two weeks; relaxin treatment using an osmotic minipump was started at week 20.
Fig. 4B is a bar graph showing the effect on the atrial remodelling (mean left atrial area) in ZSF1 rats with experimental HFpEF (n = 5-6 per group) of a continuous subcutaneous infusion of 400 pg per kg rat body weight of synthetic human relaxin-2 over two weeks; relaxin treatment using an osmotic pump was started at week 20. Fig. 4C is a bar graph showing the effect on the maximum velocity of diastolic left ventricular myocardial relaxation E’ in m/s in ZSF1 rats with experimental
HFpEF as assessed by echocardiography when rats received a daily subcutaneous injection of 100 pg human relaxin-2 per kg rat body weight or vehicle (VEH) for two weeks; daily injections were started at week 20.
Fig. 4D is a bar graph showing the effect on the E/E’ ratio in ZSF1 rats with experimental HFpEF as assessed by echocardiography when the rats received a daily subcutaneous injection of 100 pg human relaxin-2 per kg rat body weight or vehicle (VEH) for two weeks as described in Fig. 4C; daily injections were started at week 20. Fig. 4E is a bar graph showing the effect on left atrial remodeling (enlargement of left atrial area, LAA) measured as left atrial lengths (PSLAX - parasternal long axis, a specific echo view) of said groups of ZSF1 rats with experimental
HFpEF when the rats received a daily subcutaneous injection of 100 pg human relaxin-2 per kg rat body weight or vehicle (VEH) for two weeks as described; treatment started at week 20.
Fig. 4F is a bar graph comparing the lung scores of the same rats as described in Fig.
4C after the rats received a daily subcutaneous injection of 100 pg human relaxin-2 per kg rat body weight or vehicle (VEH) for two weeks, treatment started at week 20. Fig. 5a is a diagram showing a microscale thermophoresis of the high-affinity binding (KD ~ 5 nM) of human relaxin-2 to helix 12 of the ligand-binding domain of the human GC receptor in the absence of the co-activator TIF2.
Fig. 5b is a diagram of a microscale thermophoresis showing the lower affinity interaction (KD ~500 nM) of human relaxin-2 to the ligand-binding domain of the human GC receptor.
Fig. 5c is a data plot of the release of lactate dehydrogenase (LDH) from cultured primary mouse hepatocytes after induced cell injury: Control (no cell injury); induction by H2O2; induction by H2O2 after treatment of cells with relaxin-2 (Rix) or dexamethasone (Dx); after knockdown of the GC receptor by added siRNA
(GRsi) and scrambled siRNA (scr) as a control - all data in percent of detergent-induced maximal cytotoxicity. Fig. 5d is a data plot of the release of cleaved caspase-3 from cultured primary mouse hepatocytes after induced cell injury: Control (no cell injury); cell injury by H2O2; after treatment of cells with relaxin-2 (Rix) or dexamethasone (Dx); after knockdown of the GC receptor by addition of siRNA (GRsi) and scrambled siRNA (scr) as a control - all data normalized to p-actin and adjusted to the effect of H2O2.
Fig. 5e is a data plot comparing the gene expression of GC receptor (GR) in cultured primary mouse hepatocytes after induced cell injury: Control (no cell injury), by addition of H2O2; after treatment of cells with relaxin-2 (Rix) or dexamethasone
(Dx); relaxin-2 or dexamethasone alone - all data normalized to GAPDH gene expression and adjusted to control values. Fig. 5f is a data plot comparing the gene expression of mitochondrial pyruvate dehydrogenase lipoamide kinase isoenzyme 4 (PDK-4) in cultured primary mouse hepatocytes after induced cell injury: Control (no cell injury), cell injury by addition of H2O2; cell injury after treatment of cells with relaxin-2 (Rix) or dexamethasone (Dx); added relaxin-2 or dexamethasone alone - all data normalized to GAPDH gene expression and adjusted to control values. Fig. 5g is a data plot comparing concentrations of tumor necrosis factor-alpha (pg/mL) - an adipokine and cytokine - in the supernatants of activated THP1- macrophages: Control (no activation); activation by lipopolysaccharides (endotoxin); after treatment of macrophages with relaxin-2 (Rix) or dexamethasone (Dx) or Mifepristone-RU486 (RU) or LPS+Rlx+RU486 or
LPS+DX+RU486. Fig. 5h is a data plot comparing concentrations of interleukin-6 (pg/mL) - pro- inflammatory cytokine - in the supernatants of activated THP1-macrophages: Control (no activation); activation by lipopolysaccharides (endotoxin); after treatment of macrophages with relaxin-2 (Rix) or dexamethasone (Dx) or
Mifepristone-RU486 (RU) or LPS+Rlx+RU486 or LPS+Dx+RU486.
Fig. 5i is a data plot comparing concentrations of circulating tumor necrosis factor alpha (pg/mL) in blood from rats 24 hours after challenge with E. coli endotoxin:
Control (placebo - no endotoxin); challenge with E.coli endotoxin (125 pg LPS
I kg bodyweight); 2 hours after continuous sc. infusion (4 pg/h) of synthetic relaxin-2 (Relaxera Pharmazeutische GmbH, DE); i.m. injection of dexamethasone (10 mg/kg i.m.); oral RU-489 (single dose of 10 mg /kg body weight) or combinations thereof;
Fig. 5k is a data plot comparing concentrations of fasting blood sugar levels (24 hours) in rats after exposure to E. coli endotoxin: control (placebo - no endotoxin); challenge with E. coli endotoxin (125 pg LPS I kg bodyweight); challenge 2 hours after continuous subcutaneous infusion (4 pg/h) of synthetic relaxin-2
(Relaxera Pharmazeutische GmbH, Bensheim, DE); intramuscular injection of dexamethasone (10 mg/kg im.; oral RU-489 (single dose of 10 mg /kg body weight) or combinations thereof.
F ig .51 is a data plot comparing concentrations of fasting blood sugar levels (48 hours) in blood from rats after challenge with E. coli endotoxin: Control (placebo - no endotoxin); challenge with E. coli endotoxin (125 pg LPS I kg bodyweight); challenge 2 hours after continuous sc. infusion (4 pg/h) of synthetic relaxin-2 (Relaxera, Bensheim, DE); intramuscular injection of dexamethasone (10 mg/kg im); oral RU-489 (single dose of 10 mg /kg body weight) or combinations thereof.
Fig. 5m is a data plot comparing the percentage of Treg (regulatory T cells) in blood from rats after continuous sc. infusion (4 pg/h) of synthetic relaxin-2 (Relaxera) or oral RU-489 (single dose of 10 mg /kg body weight) or both - all data as a percentage of the total white blood cell count (WBC).
[0035] In Figures 5c to 5m, the boxes indicate the interquartile range, with the inner line representing the median; the whiskers indicate 1.5-times the interquartile range; values above 1.5 times (outliers) and 3 times the interquartile range (extreme range) are shown as circles and stars, respectively. Number of experiments, n = 5 for each group.
DETAILED DESCRIPTION OF THE INVENTION [0036] To date, human relaxin-2 has been administered by continuous intravenous or other infusion to ensure a consistent level of peptide in the circulation (see Scleroderma
Studies: Seibold JR et al in Safety and pharmacokinetics of recombinant human relaxin in systemic sclerosis. J Rheumatol 1998;25(2):302-7; Seibold JR et al., Recombinant human relaxin in the treatment of scleroderma. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 2000; 132(11 ):871— 9; Teerlink JR et al, in Serelaxin, recombinant human relaxin-2, for treatment of acute heart failure (RELAX-AHF): a randomised, placebo-controlled trial, Lancet (2012), http://dx.doi.org/10.1016/S0140-6736(12)61855-8; Khanna D et al in A randomized, double-Blind, placebo-controlled trial of recombinant human relaxin in the treatment of systemic sclerosis with diffuse scleroderma, Arthritis Rheum. (2009) 60(4) 1102-
1111 ; Metra M, Teerlink et al, in Effects of serelaxin in patients with acute heart failure, N Engl
J Med 2019; 381 :716-726 DOI: 10.1056/NEJMoa1801291). This mode of relaxin administration was based on a wealth of preclinical data, all obtained by continuous infusion of relaxin-2 in various animal models, mostly using osmotic minipumps (for a review, see Halls
M et al, in Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1-4, the receptors for relaxin family peptides, Pharm Rev
(2015) 67 (2) 389-440, DOI: https://doi.org/10.1124/pr.114.009472). The various human clinical trials in various cardiovascular and fibrotic diseases have been largely unsuccessful or inconclusive, failing to achieve the desired relaxin effects. [0037] In this application we disclose that more substantial relaxin effects, or at least effects like those seen after continuous administration of recombinant relaxin can be achieved by repeated boluses of synthetic human relaxin-2 at appropriate intervals. This can be achieved by daily (QD) or twice-daily (BID) subcutaneous bolus injections of small but triggering amounts of synthetic human relaxin-2 peptide, preferably as a zinc-relaxin complex, which is more stable and causes less irritation and/or inflammatory reactions at the site of injection. The efficacy of this novel route of administration has been demonstrated by an established animal model of HFpEF, the ZSF-1 rat. Here, we find already higher efficacy of bolus relaxin as compared to continuous infusion via osmotic minipumps when experimental
HFpEF is treated for only 2 weeks. The described bolus effect will significantly simplify the chronic clinical administration of relaxin-2 and improve patient compliance to such a therapeutic intervention.
EXAMPLES
EXAMPLE 1 - Pharmacokinetics of Synthetic Human Relaxin-2 After Subcutaneous Injection [0038] Healthy Sprague-Dawley rats (n = 4-8) received a subcutaneous injection of
30, 100, and 500 micrograms/kg body weight of synthetic human relaxin-2. After this sc. bolus of human relaxin-2, serum levels of synthetic human relaxin-2 were determined at predetermined intervals using an R&D human relaxin-2 ELISA (R&D Systems, Minneapolis,
USA).
[0039] The results are summarized in the plots of Figure 1 where are shown: (A) the measured serum concentration after a single subcutaneous injection of 30 micrograms/kg (squares), 100 micrograms/kg (circles) and 500 micrograms/kg (triangles) of synthetic human relaxin-2 into healthy Sprague-Dawley rats over 24 hours and (B) the same with an extended baseline of 4 hours. In summary, the pharmacokinetics following a single bolus injection of human relaxin-2 are characterized by a dose-dependent peak that occurs at after approximately 60 minutes.
EXAMPLE 2 - Pharmacokinetics of Zinc Relaxin-2 Complex after Subcutaneous Injection
[0040] Structure comparison between relaxin-2 and insulin. Relaxin is a member of the relaxin/insulin peptide hormone superfamily and is characterized by a two-chain structure constrained by disulfide bonds. The human relaxin-2 peptide can form a dimer, but the physiological role of this self-association is unclear. The solution structures of human insulin and human relaxin-2 suggest a conserved folding with local structural differences only at the peptide termini. Since relaxin-2 contains a zinc-finger structure and due to the folding similarities between insulin and relaxin-2, we tested whether human relaxin-2 and zinc ions form a stable zinc-relaxin hexamer, which we confirmed by the visible changes in solution properties and structure. This was further investigated by synthesizing various zinc-relaxin complexes as described below. The critical amino acids for the complex appear to be the tryptophanes in the B-chain of relaxin-2. Zinc-valent amino acids are found in insulin and relaxin-2 at similar structural positions. Essentially, the following amino acids within the A- and B-chains of human relaxin-2 have a potential valence for Zn(ll)-ions: L-glutamic acid, glycine,
L-histidine, L-proline, L-methionine, and L-tryptophan; see Fig. 2 A. In this study, we further investigated further various synthesized Zn-relaxin-complexes and the effect of zinc ions on serum relaxin-2 release following a subcutaneous bolus.
[0041] Synthesis of various zinc-relaxin complexes. 81.91 mg ammonium acetate (MW 77.09 g/mole) was dissolved in 100.0 mL deionized water (10.63 mM/L) at ambient temperature and adjusted to pH 5.0 with glacial acetic acid to prepare the buffer. 30 mg relaxin was dissolved in 15 mL ammonium acetate buffer, pH 5.0 to prepare the relaxin-2 acetate solution (2 mg/ml) The zinc solution was prepared by dissolving 62.83 mg zinc acetate dihydrate (MW 219.51) in 10 mL deionized water (28.63 mM/L) at ambient temperature. The relaxin-2 and Zn containing solutions at a molar ratio of 1 :1 (RLX/Zn) was prepared by mixing
2 mg of relaxin in 1 .0 mL ammonium acetate buffer (pH 5.0) with 10 pl zinc acetate solution as described. The other complexes of Zn and relaxin-2 were prepared accordingly, varying only the amount of zinc(ll) solution added, neglecting the volume added by the zinc solution. In each case, the aqueous solution of relaxin-2 immediately changed its appearance and physical behavior (fluidity) when zinc ions were added. All samples were stored at 5°C. The addition of zinc to relaxin-2 gives way to the formation of coordinated Zn-relaxin-2 complexes in which hydrophobic side groups such as the tryptophanes of the B-chain are likely buried within a multimer with increased stability and potency. [0042] Release of relaxin-2 from a zinc-relaxin-2 sc. bolus into serum. The zinc- relaxin-2 was administered to healthy Sprague-Dawley rats (n = 4 each) as a single subcutaneous bolus injection at 1 mg/kg bw. Relaxin serum levels were determined again using a R&D human relaxin-2 ELISA (R&D Systems, Minneapolis, USA). The results have been summarized in Fig. 2B. Blue (triangels) synthesized human relaxin-2 as reference; orange (light full circles) Zn-relaxin complex at molar ratio relaxin: Zn of 1 :1 ; gray (medium full circles), relaxin: Zn 1 :2; yellow (light gray circles), relaxin: Zn 1 :4.
[0043] As can be seen from Fig. 2B, a subcutaneous bolus of the zinc(ll) relaxin complex results in each case in a steadier release of relaxin-2 into the serum over a longer period of time and a later and lower peak serum relaxin bolus. Serum relaxin bolus peaks were observed between 180 and 240 minutes instead of 60 minutes as observed with pure relaxin-
2, which represents a tremendous pharmacological advantage for therapeutic inventions. The more gradual release of relaxin-2 with a subcutaneous injection of zinc-relaxin-2 complex results in a longer bolus persistence in the circulation for the desired therapeutic interventions.
In addition, the zinc-relaxin complex results in a lower osmolality, allowing for highly concentrated subcutaneous formulations with little or no irritation and local inflammation at the site of injection. A clinical option for subcutaneous injection should be isotonic and isosmotic with non- or minimally antigenic active agents. [0044] With respect to the formulation of the “zinc relaxin-2-bolus” or “pure relaxin-2 bolus”, carbohydrates may be added to adjust the osmolality of the formulation for subcutaneous injection. Ideally, the relaxin medication should be formulated as an isotonic solution (osmolality of about 300 mOsm/kg and no more than 600 mOs/kg) in order to prevent pain. A pH close to the physiological one is recommended to minimize pain, irritation, and tissue damage. Cyclodextrins may be used as complexing agents to increase the aqueous solubility of the complex and in order to increase its bioavailability and to improve stability. Cyclodextrins can lowerthe free concentration of the medication in the circulation and therefore the pharmacokinetics/pharmacodynamics may be changed significantly. In addition, cyclodextrins, dextrins and carbohydrates such as mannitol can be used to reduce or prevent irritation at the injection site by adapting the osmolality of the medicinal formulation.
Cyclodextrins (CDs) are cyclic oligosaccharides made up of a number of dextrose units of (a- 1 ,4)lin ked a-D-glucopyranose. These cyclic structures contain a lipophilic central cavity and a hydrophilic outer surface. Cyclodextrins are made up of six, seven or eight dextrose units (a-,
P-, and yCDs, respectively; the so-called parent cyclodextrins. Ph.Eur names of a- and p-CD: alfadex and betadex. Cyclodextrins interact with molecules to form inclusion complexes and can be used e.g. to improve the aqueous solubility of the active ingredient. For P-CD, which itself has a relatively low aqueous solubility, substitution of any of the hydrogen bond-forming hydroxyl groups, even by lipophilic functions, results in a dramatic improvement in the aqueous solubility of the derivative. Examples of p-CD derivatives used as excipients in medicines are the sulfobutylether of PCD (SBE-P-CD), the hydroxypropyl derivative of p-CD (HP-P-CD, hydroxypropylbetadex), and the randomly methylated p-CD (RM-P-CD). EXAMPLE 3 - Effectiveness of Relaxin-2 using a Continuous Subcutaneous Infusion [0045] Besides the eNOS-knockout mouse on high-fat diet (Schiattarella G.G. et al. in
Nitrosative stress drives heart failure with preserved ejection fraction Nature 2019, 568: 351- 356), the ZSF1 rat model for experimental HFpEF is the most established animal model to date for studying heart failure with preserved ejection fraction (Hamdani N et al in Myocardial Titin Hypophosporylation importantly contributes to Heart Failure with Preserved Ejection Fraction in a Rat Metabolic Risk Model, Circulation Heart Failure (2013) 6(6): 1239-1249; Schauer A et al., in ZSF1 rat as animal model for HFpEF: Development of reduced diastolic function and skeletal muscle dysfunction, ESC Heart Failure 2020, 7: 2123-2134). This is because the ZSF1 rat model of HFpEF best mimics the multiple human drivers of human HFpEF, i.e. the fact that in HFpEF develops in humans against a background of many coexisting pathogenetic factors such as hypertension, obesity and metabolic inflammation, diabetes, lung disease. This animal model was developed by crossing rat strains with two separate leptin receptor mutations (fa and facp), the lean female ZDF rat (+/fa) and the lean male SHHF rat (+/facp). Offspring homozygous for both mutations (fa:facp) are obese and develop insulin resistance, hyperglycaemia and mild hypertension (ZSF1 -obese). The heterozygous offspring (ZSF1-lean) are lean and show no signs of obesity or diabetes.
[0046] Table 1 describes the development of diastolic dysfunction in male ZSF1 -obese rats (n = 8 ZSF1 -obese males) fed a high fat diet from 10 weeks to 18 weeks of age. The animals have preserved left ventricular ejection fraction, but reduced global longitudinal strain (GLS), which closely mimics the pathological situation in human HFpEF patients.
Table 1
Diastolic dysfunction in ZSF1 rats as assessed by echocardiography
Figure imgf000018_0001
Figure imgf000018_0002
Figure imgf000018_0003
EDVI: end-diastolic volume index; Ejection Fraction: left ventricular ejection fraction; E‘: maximum velocity of diastolic left ventricular myocardial relaxation in m/s); AET: Aortic Ejection Time; MV E/E‘: a dimensionless measure of left ventricular compliance (E, maximum velocity of mitral diastolic inflow in m/s; E‘, maximum velocity of diastolic left ventricular myocardial relaxation in m/s); GLS: Global Longitudinal Strain. [0047] Diminished Global Longitudinal Strain (GLS) is a well-established myocardial deformation analysis that predominantly reflects the function of subendocardial longitudinally oriented fibres, which are most susceptible to ischaemic damage and wall stress. Therefore, they may exhibit abnormal contraction patterns in the setting of an apparently normal left ventricular ejection fraction (LVEF). The E/E’ criterion also defines diastolic dysfunction in patients; its increase in absolute terms is mainly due to an impaired rate of myocardial relaxation (E’). In addition, the left atrial enlargement (see left atrial area) is a key feature of structural remodelling due to reduced left ventricular compliance. In combination with general clinical signs of heart failure as pulmonary oedema (the Lung Score), this detection of diastolic dysfunction completes the diagnostic criteria for HFpEF.
[0048] Using this animal model, ZSF1 rats (n = 8-12 per group) with experimental
HFpEF then received synthetic human relaxin-2 to assess the effects of relaxin-2. In brief, nine-week old ZSF1 lean rats were obtained from Charles River and fed with Purina Diet (#5008). After a 1-week laboratory adaptation period, animals underwent phenotypic evaluation and echocardiographic evaluation. From this point onward, the group of ZSF1 obese rats was allocated to receive HFD (Research Diet Inc, #D12468). To assess diastolic function, peak velocity of early (E) and late (A) mitral inflow signals and the ratio of E over e'
(peak velocity of early diastolic lateral mitral annular motion) were measured as an indication of LV filling pressure. More precisely, E/E‘ is a dimensionless measure of left ventricular compliance (E, maximum velocity of mitral diastolic inflow in m/s; e‘, maximum velocity of diastolic left ventricular myocardial relaxation in m/s). Furthermore, the Left Atrial Area was determined which reflects left atrial remodeling (enlargement) attributable to impaired left ventricular filling characteristics (impaired compliance). At 20 weeks, animals further underwent hemodynamic evaluation and biomechanical studies under anesthesia.
Investigation conformed to the Guide for the Care and Use of Laboratory Animals published by the National Institutes of Health (NIH Publication no. 85-23, revised 1996); cf. Figs. 3A-D. [0049] Treatment with relaxin started at week 20, and relaxin-2 was compared to placebo to see whether relaxin-2 can significantly improve in rat the above echocardiography parameters of diastolic dysfunction, which are also important in the clinical assessment of human HFpEF.
[0050] Results: The effect of the continuous infusion of 400pg per kg rat body weight per day over two weeks of human relaxin-2 using an osmotic minipump in these rats with experimental HFpEF have been summarized in Fig. 4A and Fig. 4B The data support that the continuous administration of relaxin-2 has a substantial effect on the echocardiographic parameter (E/e’) of diastolic dysfunction as well as on the Left Atrial Area, which - most importantly - are also crucial for the clinical assessment of human HFpEF. Statistical analyses were performed using a two-way nonparametric ANOVA followed by Mann-Whitney U test for ranks. *, P < 0.05; **, P < 0.01 ; ***, P < 0.001 ; ****, P < 0.0001 .
Figure imgf000020_0001
[0051] The same animal model was used as in Example 3, but in this 2-week comparative experiment, the human relaxin-2 was administered as a daily subcutaneous bolus of 100 micrograms/kg bodyweight, starting at week 20. Figs. 4C-4F summarise the effect of
2 weeks treatment of ZSF1 rats (n = 5-6 per group) with daily subcutaneous boluses of 100 micrograms/kg body weight of synthetic human relaxin-2. As with continuous infusion, this relaxin drug significantly improves diastolic dysfunction in this HFpEF animal model compared to placebo injections. This includes E and E/e‘ as a echocardiographic parameter of left ventricular compliance and left atrial length measured in PSLAX (parasternal long axis, a specific echo view) as measure of atrial remodeling and also the Lung Score. Statistical analyses were performed using a two-way non-parametric ANOVA followed by Mann-Whitney
U test for ranks. *, P < 0.05; **, P < 0.01 ; ***, P < 0.001 ; ****, P < 0.0001 .
[0052] It is noteworthy, that the total amount of relaxin-2 drug administered by the daily subcutaneous bolus was only a quarter of the amount of relaxin-2 administered by the comparative continuous infusion as prescribed in the prior art. However, the essential therapeutic effect was the same, if not statistically greater. Consequently, this suggests that the therapeutic effects of relaxin-2 do not depend on a momentary serum concentration, but that the relaxin activity appears to affect mechanisms at the DNA level. In this context, it should not been overlooked that relaxin-2 appears to act on the glucocorticoid receptor and on the canonical Wnt signaling, where relaxin-2 seems to able to by-pass the effects of Dickkopf proteins in serum. Without wishing to be bound by theory, the present inventors assume that the reversal of myocardial ageing by relaxin-2 is mediated by the reactivation of canonical Wnt signaling, which primarily requires initiation rather than a constant trigger.
[0053] The use of zinc complexes of relaxin-2 further result in a flattened and prolonged pharmacokinetic curve with delayed peak levels. Their use may prolong individual bolus persistence in circulation and thereby confer another degree of freedom to optimally adjust the described bolus administration for clinical studies or therapeutic interventions. EXAMPLE 5 - Effects of Relaxin-2 on the Glucocorticoid Receptor
[0054] The new dosis regimen and mode of application for HFpEF patients affect also the treatment other diseases. Millions of patients take glucocorticoids to treat autoimmune and rheumatoid diseases, neurological disorders, pulmonary diseases, cancer, and other diseases and causes. However, the chronic side effects of glucocorticoids and their adverse consequences are much feared, particularly the negative impact due to glucocorticoid receptor (GR) down-regulation, steroid-induced hyperglycemia, or activation of gluconeogenesis as well
1 as Cushing’s syndrome. The present inventors have discovered that relaxin-2 binds the ligand- binding domain of the GR like steroids and glucocorticoids, forming an activated GR-ligand complex. The relaxin-GR complex activates the transcription of genes dampening the innate immune system but no genes activating gluconeogenesis, unlike the glucocorticoids or corticosteroids. This discovery enlarges the therapeutic applications of relaxin-2 for patients in need of a dampened innate immune system. This group of patients comprises for example patients who receive or have received an allotransplant, and cancer patients. Another large group are patients suffering from forms of tissue/endothelial injury or tissue-damaging diseases, including autoimmune or rheumatic tissue-damaging diseases comprising ankylosing spondylitis (AS) and spondylarthritis, fibromyalgia, gout, infectious arthritis, lupus, systemic autoimmune disease, osteoarthritis (OA), psoriatic arthritis (PsA), and inflammatory types of arthritis, rheumatoid arthritis (RA). Medications for these diseases include corticosteroids, oral and topical analgesics, non-steroidal anti-inflammatory drugs such as ibuprofen and COX-2 inhibitors, and disease-specific biologies. Tissue injury, endothelial injury, and endothelial cell activation, particularly in allograft rejection, can be detected and monitored by increased concentrations of calprotectin and/or S100A12 in extracellular fluids and the bloodstream. This is because endothelial cells also play a critical role in immune cell recruitment and extravasation. The calcium-binding S100 proteins, particularly calprotectin and S100A12, have a wide range of intracellular and extracellular functions, including regulation of calcium balance, cell apoptosis, cell migration, differentiation, proliferation, energy metabolism, and inflammation. The calcium-binding S100 proteins are released from the cytoplasm of the endothelial cell when triggered by tissue/cell damage, antibody stress, and endothelial stress.
The S100 proteins then serve as danger signals, DAMP (damage-associated molecular pattern) molecules, and are involved in the regulation of immune homeostasis (macrophage migration, invasion, and differentiation), post-traumatic injury, and inflammation. They are therefore biomarkers in some specific diseases such as IBD (inflammatory bowel disease), although their multiple functions must be assigned to cell migration, differentiation, tissue repair, immune homeostasis, and inflammation management. The lack of commonly available diagnostic tests for tissue injury, endothelial injury, endothelial stress, and anti-endothelial cell antibody binding, which undoubtedly lead to allograft dysfunction and allograft rejection, makes calcium-binding S100A12 and calprotectin biomarkers for endothelial activation, immune cell recruitment, endothelial injury, endothelial antibody binding, and complement activation.
[0055] The present inventors discovered that relaxin-2 not only binds to the glucocorticoid receptor but that the experimental results further indicate that the administration of relaxin-2 leads to a specific activation and promotion of regulatory T cells at local and systemic levels, likely induced by the relaxin-GR complex. This allows suppression of immune responses and significantly widens the use of relaxin-2 as an active ingredient in pharmaceuticals for treating abnormal, excessive, and undesired tissue-damaging immune responses to self- and foreign antigens. While the promotion of peripherally induced Treg cells could also be achieved by the administration of glucocorticoids, such treatment is disadvantageous due to the Cushingoid adverse effects of glucocorticoids, corticosteroids, and synthetic analogs thereof. Their adverse effects are well-known and numerous (cf. 2022 ICD-
10CM Code T38.0X5A).
[0056] Regulatory T cells (Treg cells) were originally defined as CD4+ T cells with a high expression of CD25 (interleukin-2 receptor a-chain). The regulatory T cells are further classified into thymic and peripherally induced Treg cells based on where they develop. The
Foxp3 gene, a member of the Forkhead/winged-helix family of transcriptional regulators, was discovered to be an important regulator in the development of Treg cells based on the following findings: Scurfy mice with a frameshift mutation in the Foxp3 gene have T cell inflammation in multiple organs and a lethal autoimmune disease due to effectorT cell activation and increased cytokine production caused by the absence of Treg cells. In addition, mutation of the Foxp3 gene in humans leads to IPEX syndrome (X-linked immune dysregulation, polyendocrinopathy, and enteropathy). In addition, forced expression of FoxP3 in naive T cells leads to immune suppressive function. CD4/CD25-naive T cells transfected with the Foxp3 gene can transform into CD4+CD25+ Treg-like cells that produce inhibitory cytokines and express typical Treg-cell molecules such as CD25, cytotoxic T-lymphocyte antigen-4 (CTLA-4), and glucocorticoid- induced tumor necrosis factor (TNF) receptor-related protein (GITR). Thus, FoxP3 is a lineage- specific marker and an important regulatory gene for the generation, maintenance, and immune suppressive functions of Treg cells. Regulatory T cells are required to suppress abnormal or excessive immune responses and to maintain homeostasis and self-tolerance by inhibiting T cell proliferation and cytokine production. The Treg cells exert their immunosuppressive function through dominant consumption of the cytokine interleukin-2 and by inhibitory cytokines (TGF-B, IL-10, IL-35) as well as induction of apoptosis or a killing of effector or antigen-presenting cells (APC) by perforin, granzyme B or Fas ligand interaction.
The other immunosuppressive mechanisms of Treg cells involve immune checkpoint molecules and include inhibition of effector T cells by the lymphocyte activation programmed cell death pathways or the cytotoxic T-lymphocyte antigen (CTLA-4). A third immunosuppressive mechanism may be metabolic modulation by indoleamine 2,3- dioxygenase (IDO) expression, which affects the kynurenine-tryptophan pathway in dendritic cells. The Treg cells thereby play a critical role in suppressing autoimmunity and inflammation.
Reduced number and function of Treg cells are associated with human autoimmune disease, and activation and augmentation of Treg cells have been shown to be beneficial in treating autoimmune diseases in clinical trials (see the review of Margarita Dominquez-Vallar & David A. Hafler, Regulatory T cells in autoimmune disease, Nature Immunology 2018, 19, 665-673).
Overall, all current results suggest that Treg cells contribute to the maintenance of self- tolerance by downregulating the immune response to self and foreign antigens in an antigen- nonspecific manner Therefore, it is reasonable to hypothesize that a relaxin-2-induced increase in Treg cells in a posttransplant patient also improves post-transplant outcomes beyond the prevention of ischemic injury. The very same can also be assumed when relaxin-2 is used in treating autoimmune induced tissue injury, endothelial cell injury and diseases following endothelial cell activation. The hypothesis is also supported by the fact that a
1 i reduction in the proportion of Treg cells in the peripheral blood is known to lift general immune suppression, thereby enhancing the innate and acquired immune response to foreign and self- antigens. Therefore, the present discovery significantly expands the pharmaceutical toolbox.
EXAMPLE 5 Relaxin binds and activates the glucocorticoid receptor
[0057] Fig.5a and Fig. 5b refer to in vitro binding and affinity studies of synthetic human relaxin-2 (shRIx) to the ligand-binding domain of the glucocorticoid receptor (GR-LBD) using microscale thermophoresis (MST). The microscale thermophoresis is based on measuring the directed movement of molecules in localized temperature gradients created by IR laser radiation in high-precision glass capillary tubes containing the interacting partners - synthetic human relaxin-2 and recombinant GR-LBD. For this experiment, the human glucocorticoid receptor (GR-LBD) ligand-binding domain was expressed in an E. coli expression system to obtain a large amount of soluble protein stable for biophysical characterization. The recombinantly produced GR-LBD showed little aggregation and proved to be fully functional. One of the interacting partners was labeled with a fluorescent dye and added to a serial dilution series (15 dilutions each) of the non-fluorescent partner. After incubation, the thermophoretic movement of the complex is detected. Conformational changes due to ligand binding to the target or binding near the fluorophore induce thermophoretic changes. The affinity of interacting protein is determined by analyzing the change in normalized fluorescence as a function of the concentration of titrated binding partner. The next step was to determine the binding mode of relaxin and the activation mechanism of GR. Fluorescence polarisation revealed two binding affinities in the pico- and nanomolar range. Moreover, human
H2 relaxin could displace fluormone labeled GS red out of its binding pocket on GR-LBD (see method described by Hemmerling M et al. in Selective Nonsteroidal Glucocorticoid Receptor
Modulators for the Inhaled Treatment of Pulmonary Diseases, J. Med. Chem. 2017, 60, 20, 8591-8605). Using this combination of biophysical and structural biology techniques, including microscale thermophoresis (MST), hydrogen-deuterium exchange mass spectrometry (HDX- MS), and NMR, the relaxin-2 binding site of the glucocorticoid receptor was identified, which was the steroid-binding pocket of the GR-LBD.
[0058] The effects of relaxin binding on GR-LBD have further been investigated to determine whether relaxin binding activates the receptor like an agonist or represses transcriptional activity by acting like an antagonist. GR-LBD has an activation function-2 site that will recruit cofactors upon ligand binding. The cofactors (coactivators or corepressors) are specific to the cellular environment. Therefore, the binding of relaxin with coactivator and corepressor motifs to the GR-LBD/relaxin complex was tested. [0059] It was found that relaxin binds to both cofactors, resulting in different receptor conformational changes. Since thermophoresis is an intrinsic phenomenon of a molecule that depends on the hydration shell and the size, the binding events could be identified by tracking the associated changes to thermophoresis in a fluorescently labeled interacting partner. In summary, the results of the microscale thermophoresis (see Figs. 5a and 5b) indicate a high- affinity interaction (KD ~ 5 nM) and another lower-affinity interaction (KD ~ 500 nM) of synthetic human relaxin-2 with the GR-LBD.
[0060] According to hydrogen-deuterium exchange experiments (not shown), human relaxin-2 appears to bind to helix 12 of the LBD, but in contrast to classical glucocorticoids in absence of the transcription co-regulator NCoA-2 (nuclear receptor coactivator 2). NCoA- 2 is also known as glucocorticoid receptor-interacting protein 1 (GRIP1), steroid receptor coactivator-2 (SRC-2), or transcription intermediary factor 2 (TIF2). NCoA-2 contains several nuclear receptor interacting domains and intrinsic histone acetyltransferase activities, and when GR recruits NCoA-2 to a DNA promotion site, the role of NCoA-2 also appears to be in acetylating histones so that the downstream DNA becomes more accessible for transcription.
The presence and amount of NCoA-2 are cell-type dependent. NCoA2 (GRIP1 , SRC-2, TIF2) therefore supports up-regulation of DNA expression which also leads to increased activation of genes responsible for gluconeogenesis. Since human relaxin-2 does not recruit NCoA-2 upon binding, this type of gene activation does not appear to be triggered when human relaxin-
2 binds to the glucocorticoid receptor. Relaxin-2 activates transcription of genes dampening peroxide-induced cytotoxicity, inflammatory responses, and apoptosis
[0061] With reference to Fig. 5c and Fig. 5d, mouse hepatocytes were isolated as described by Tamaki N et al. in Am J Physiol Gastrointest Liver Physiol 2008294, G499. Briefly, livers from mice anesthetized with pentobarbital were washed and perfused for 5 min with buffer consisting of (all values are mg/L) 8,000 NaCI, 400 KCI, 88.7 NaH2PO4 H2O, 120.45
Na2HPO4, 2,380 HEPES, 350 NaHCO3, 190 EGTA, and 900 glucose, pH 7.25, then treated with 0.03% collagenase at 37°C for 15 min in digestion buffer containing (all values are mg/L)
8,000 NaCI, 400 KCI, 88.7 NaH2PO4 H2O, 120.45 Na2HPO4, 2,380 HEPES, 350 NaHCO3, and
560 CaCl2'2H2O , pH 7.25. After collagenase perfusion, the liver capsule was isolated, and the cells dispersed in Geys balanced salt solution (GBSS)-B consisting of (all values in mg/L) 8,000
NaCI, 370 KCI, 210 MgCI2'6H2O, 70 MgSO4-7H2O, 120 NaH2PO4, 30 KH2PO4, 991 glucose,
227 NaHCO3, and 225 CaCl2'2H2O (pH 7.25). Cells were additionally separated by forcing the material through a steel mesh and collecting the cells by centrifugation at 50 G for 1 min. The cell pellet was resuspended in GBSS-B solution and washed three times with intermittent centrifugations.
[0062] Isolated mouse hepatocytes were cultured on type I collagen-coated 6-well plates coated at a cell density of 5 x 105 cells/well with Dulbecco's modified Eagle's medium containing 10% fetal calf serum, 100 U/ml penicillin, and 100 pg/ml streptomycin at 37°C in a humidified atmosphere of 5% CO2-95% air. After plating, the medium was replaced with serum- free Dulbecco's modified Eagle medium after 6 hours. The hepatocytes were then subjected to hydrogen peroxide treatment (2 mM H2O2/L) for 5 hours with or without pretreatment with synthetic human relaxin-2 (10 nM/L, 24 h) (Relaxera Pharmazeutische GmbH, Bensheim, DE) or dexamethasone (0.5 mM, 24 h) (Sigma Aldrich). In addition, hepatocytes were also transfected with GR-siRNA or scrambled siRNA using lipofectamine reagent (Invitrogen) to test whether the release of LDH or caspase-3 into the medium in these two experiments was dependent on the ligand-activated GR (n = 5 for each group). H2O2-induced cell injury
(cytotoxicity) was determined immunologically by quantification of lactate dehydrogenase (LDH) released into the culture medium using an enzyme-linked assay according to the manufacturer’s instructions (Goat LDH ELISA kit, Biomol Feinchemikalien GmbH, DE) and by
Western Blot analysis of cleaved caspase-3 (Caspase-3 Rabbit mAb #14220, Cell Signaling
Technology, Danvers, MA, US). [0063] The results are summarized in Figs. 5c and 5d. LDH release data shown in
Fig. 5c are expressed as a percentage of detergent-induced maximum cytotoxicity. Data on cleaved/activated caspase-3 shown in Fig. 5d were normalized to p-actin and adjusted for the effect of H2O2 (* p < 0.05 vs. control; #, p < 0.05 vs. H2O2; Kruskal-Wallis ANOVA on ranks for global testing with post hoc Mann-Whitney U-tests for pairwise comparisons (Bonferroni-Holm adjustment of p). In summary, Figs. 5c and 5d show that both relaxin-2 (Rix) and dexamethasone (Dx) can markedly dampen peroxide-induced cell injury (LDH release) and apoptosis (cleaved caspase-3) and that this attenuating effect does not occur when the expression of GR is specifically knocked out by GR-siRNA, whereas a knock-out using scrambled siRNA (scr-siRNA) showed no effect. [0064] Physiologically, LDH is an enzyme expressed in nearly all living cells, including heart muscles and blood cells, and it catalyzes the conversion of lactate to pyruvate and back.
Because it is released during tissue injury, it is a marker of common injuries, damaged tissues, and diseases involving tissue damage such as heart failure. In contrast, the relative concentrations of its substrates primarily regulate LDH activity. LDH is subject to transcriptional regulation by peroxisome proliferator-activated receptor-y coactivator 1a (PGC-1a) in an estrogen-related receptor-a-dependent manner. [0065] Caspase 3 protein (CASP3) or cysteine-dependent aspartate-directed protease 3 plays an essential role in programmed cell death. Caspase-3 is synthesized as an inactive zymogen until cleaved after apoptotic signaling events. Caspase-3 is thought to ensure that cellular components are degraded in a controlled manner, and that cell death occurs with minimal impact on surrounding tissues. Caspase deficiency has been identified as a cause of tumor development, for example, by a mutation in a cell cycle gene that removes cell growth restrictions in combination with mutations in apoptotic proteins such as caspases that would trigger cell death in abnormally growing cells. Conversely, over-activation of caspase-3 can lead to excessive programmed cell death. This is seen in several neurodegenerative diseases in which neural cells are lost, such as Alzheimer's disease. Caspases involved in processing inflammatory signals are also implicated in disease. Insufficient caspase activation can increase an organism's susceptibility to infection because an appropriate immune response may not be triggered. For example, inflammatory caspase-1 has been linked to the development of autoimmune diseases; drugs that block caspase activation have been used to improve patients’ health.
[0066] In conclusion, synthetic relaxin-2 as a ligand of the glucocorticoid receptor appears to have cell biological effects like dexamethasone in attenuating an inflammatory response to tissue injury as well as apoptosis and necrosis.
Relaxin-GR complex does not activate transcription of genes involved in gluconeogenesis [0067] Mouse hepatocytes were obtained and tested for glucocorticoid-adverse effects after treatment with synthetic human relaxin-2 or dexamethasone (500 nM, 24 hours). The complex physiological adverse effects of glucocorticoid excess are numerous and difficult to assess (Cushing’s syndrome, diabetes, skin thinning, hypertension, osteoporosis, obesity, impaired wound healing, depression, etc.), but measurable in this cell assay is the activation of genes involved in gluconeogenesis and in diabetes, obesity, and impaired wound healing. Consequently, the regulation of GR- and PDK-4 transcription was examined by qRT-PCR in normal primary mouse hepatocytes for control and in H2O2-stressed primary mouse hepatocytes from example 2 after 24 hours of treatment with 10 nM/L synthetic human relaxin-
2 (Relaxera Pharma. GmbH&Co.KG) and 500nM/L dexamethasone (Sigma Aldrich). Results are shown in the block diagrams of Figs. 5e and 5f.
[0068] Specifically, Fig. 5e shows that relaxin-2 increases GR gene transcription in normal and H2O2-stressed primary mouse hepatocytes by 100% to 200 %, whereas dexamethasone has no such effect. Fig. 5f shows that the incubation with relaxin-2 has no effect on PDK-4 gene transcription in normal and H2O2-stressed primary mouse hepatocytes. However, when primary mouse hepatocytes are incubated with dexamethasone, PDK-4 gene transcription increases severalfold. The combination of Figs. 5e and 5f is therefore strong evidence that the complex of relaxin and GR binds to a genomic DNA locus that is different from the DNA locus of steroid-activated GR.
[0069] Physiologically, PDK-4 (pyruvate dehydrogenase lipoamide kinase isozyme 4) is a mitochondrial protein that inhibits the pyruvate dehydrogenase complex (PDH) by phosphorylating one of its subunits. An active PDH complex is required to convert pyruvate to acetyl-CoA for the glycolytic products to enter the citric acid cycle. Fasting results in an induction of PDK-4 mRNA and PDK-4 enzyme in both cardiac and skeletal muscle, suppressing glucose oxidation during starvation as part of an integrative response and for glucose maintenance. The PDK-4 enzyme is therefore thought to play a critical role in regulating glucose metabolism, whereas an increased PDK-4 transcription indicates gluconeogenesis. It is well known that PDK-4 expression is physiologically regulated by glucocorticoids, retinoic acid, and insulin which enhance the transcription of the PDK-4 gene in white adipose tissue. Oxidation of fatty acids is also increased when the PDK-4 level is elevated. Insulin downregulates PDK-4 mRNA transcription. When cells are exposed to dexamethasone to increase PDK-4 mRNA expression, insulin blocks this effect and the oxidation of fatty acids. In type 2 diabetes, PDK-4 is overexpressed in skeletal muscle, resulting in impaired glucose utilization. In patients suffering from obesity, PDK-4 mRNA expression is also markedly decreased in association with increased glucose uptake, likely due to the downregulation of PDK-4 by insulin. This is consistent with the hypothesis that fatty acid availability affects glucose metabolism by regulating the pyruvate dehydrogenase (PDH) complex. Indeed, in insulin-resistant individuals, an inadequate downregulation of PDK-4 mRNA may cause increased PDK-4 expression, leading to impaired glucose oxidation followed by increased fatty acid oxidation. Conversely, PDK-4 is downregulated in cardiac muscle tissue during heart failure, which is a physiological countermeasure (Razeghi P et al., in
Downregulation of metabolic gene expression in failing human heart before and after mechanical unloading, Cardiology 2002, 97(4):203-9).
[0070] The results in Figs. 5c through 5f show that relaxin-2 like glucocorticoids, corticosteroids, and mineralocorticoids has anti-inflammatory effects, but without leading to impaired glucose oxidation, increased fatty acid oxidation, and gluconeogenesis. The ubiquitous role of PDK-4 further suggests that a pharmaceutical composition containing relaxin-2 and treatment with relaxin-2 may be an alternative to glucocorticoid treatment because it results in increased expression of the glucocorticoid receptor, which suppresses immune and inflammatory responses, but not in increased expression of PDK-4, which is unfavorable for glucose metabolism and balance.
[0071] The glucocorticoid receptor (GR) is an evolutionarily conserved ligand- dependent transcription factor. Upon binding of a steroid hormone or other ligand, the receptor migrates from the cytoplasm to the nucleus where it binds to a genomic DNA locus and positively or negatively modulates the transcription rates of the locus-associated genes.
Tremendous efforts have been made to uncover the molecular signaling actions of the GR, including intracellular shuttling, transcriptional regulation, and interaction with other intracellular signaling pathways. In brief, glucocorticoids are essential for the maintenance of resting-state and stress response and, therefore, they are essential in the treatment of numerous diseases, including autoimmune, inflammatory, allergic, and lymphoproliferative disorders. The pathological or therapeutic implications of the GR cannot be overstated. These include, except for genetic alterations in the human GR gene, disease-associated GR regulatory molecules and the development of GR ligands with selective GR action. [0072] Figs. 5a to 5f show that the administration of synthetic relaxin-2 has therapeutic effects that differ from known corticosteroids and glucocorticoids. The experiments presented demonstrate that synthetic relaxin-2 elicits GR-dependent glucocorticoid effects, including inhibition of apoptosis in mouse hepatocytes and inhibition of cytokine release in human macrophages (see example 4 below), which, in contrast to classical corticosteroids and glucocorticoids, avoid unwanted effects such as GR down-regulation or glucocorticoid- or steroid-induced hyperglycemia or activation of gluconeogenesis. Relaxin-2 dampens the release of pro-inflammatory cytokines [0073] THP-1 cells were differentiated into macrophages and cultured as described by Dschietzig T et al. in Identification of the pregnancy hormone relaxin as a glucocorticoid receptor agonist, FASEB J 2004, 18:1536-1538. Briefly, the THP-1 cells are derived from a cell line generated from a human monocyte leukemia, and when the cells are treated in the passage with myristate-phorbol ester, they differentiate into macrophages as is well known in the art. [0074] Referring to Figs. 5g and 5h, the macrophages were then challenged for 24 hours with 10 ng/ml Salmonella abortus equii endotoxin (Sigma Aldrich) in the presence or absence of synthetic human relaxin-2 (10 nM/L) and dexamethasone (500 nM/L) and/or the
GR antagonist RU-486 (500 nM) (Sigma Aldrich), or combinations thereof (n = 5 for each group). RU-486 binds to the GR in the steroid pocket of the ligand-binding domain.
Subsequently, the supernatant levels of TNF-a and interleukin-6 were determined by ELISA
(R&D Systems). [0075] The results are summarized in the block diagrams for Figs. 5g + h, and demonstrate that the endotoxin induced these macrophages to produce and excrete the proinflammatory cytokines tumor necrose factor alpha (TNF) and interleukin-6 (IL-6), which inflammatory response was both depressed in the presence of synthetic human relaxin-2 and dexamethasone. This response can be classified as GR-dependent, as RU-486 completely inhibited this effect. [0076] As further background, RU-486, also known as Mifepristone®, is a steroidal antiprogesterone as well as an antiglucocorticoid and antiandrogen. It competitively antagonizes cortisol action at the GR receptor. In humans, an antiglucocorticoid effect of RU-
486 is observed at doses greater or equal to 4.5 mg/kg through a compensatory increase in adrenocorticotropic hormone (ACTH) and cortisol. In animals, a weak antiandrogenic effect is seen with prolonged administration of very high doses (Danco Laboratories, 2005, Mifeprex
U.S. prescribing information). Thus, this example demonstrates that the administration of synthetic human relaxin-2 can provide GR-mediated regulation of the immune system, and has immune-suppressive properties, but without the side effects of inducing gluconeogenesis and insulin insensitivity.
Relaxin-GR complex does not induce hyperglycemia other than to dexamethasone-GR [0077] Figs. 5i, 5k, and 5L refer to animal experiments and show the different effects of relaxin-2 and dexamethasone (Sigma Aldrich) administration on blood glucose levels in rats after 24 and 48 hours. In brief, male and female Sprague-Dawley rats (body weight 300 - 350 g) were treated with an intraperitoneal injection of E. coli endotoxin (125 pg/kg body weight) or placebo (vehicle). Blood from the tail vein was taken after 24 hours for the determination of circulating TNF-a (ELISA, R&D Systems) and fasting blood glucose and at 48 hours for measurement of fasting blood glucose. Two hours before administration of endotoxin or placebo, animals received dexamethasone (intramuscular 10 mg/kg body weight), synthetic relaxin-2 (Relaxera Pharmazeutische GmbH&Co.KG, Bensheim) as a subcutaneous infusion (4 pg shRIx/h) over 12 hours via osmotic Alzet minipumps, oral RU-486 (single dose of 10 mg/kg body weight), or combinations thereof (n = 5 animals per group). The results are shown in Figs. 5k and 5L; *, p < 0.05 vs. control; #, p < 0.05 vs. endotoxin plus dexamethasone;
Kruskal-Wallis ANOVA on ranks for global testing with posthoc Mann-Whitney U-tests for pairwise comparisons (Bonferroni-Holm adjustment of p).
[0078] While dexamethasone markedly enhanced the endotoxin-related increase in blood glucose, reflecting the animal’s disease and hyperthermia response to endotoxin, relaxin-2 decreased glucose levels compared with endotoxin alone. This was true at both 24 and 48 hours after endotoxin. Oral administration of RU-486 inhibited the effects of relaxin-2 and dexamethasone by its antagonistic actions at the glucocorticoid receptor, as also the concentration of TNF-a in the circulation. In conclusion, human relaxin-2 attenuated the endotoxin-induced surge of circulating TNF-a in rats of both sexes. The effect could be identified as GR-dependent and comparable to that of dexamethasone, a classic glucocorticoid. However, unlike dexamethasone, relaxin-2 did not induce hyperglycemia which is a medically extremely important finding.
Relaxin-2 furthers differentiation of naive T cells into regulatory T cells (Treg) in mice
[0079] Mice of C57BI/6 background (each groups n=5) were intraperitoneally injected once daily for 3 consecutive days in the following experimental groups: synthetic human relaxin-2 (Relaxera) (10 micrograms/kg body weight), placebo (vehicle, sodium acetate), RU- 486 (2.5 mg/kg body weight), relaxin-2 plus RU-486. Thereafter, mice were sacrificed, their spleens were processed using standard procedures, and the percentage of spleen regulatory T cells (Treg) was analyzed by FACS. Treg cells were originally defined as characterized as CD4+FoxP3+ cells wherein FoxP3 (a Forkhead transcription factor) is the Treg master regulator. Regulatory T cells expressing the transcription factor Forkhead box P3 (FoxP3) are known to control immune responses and prevent autoimmunity.
[0080] As shown in Fig. 5m the administration of relaxin-2 approximately doubled the percentage of regulatory T cells (CD4+FoxP3+ cells) as compared with placebo. The GR antagonist RU-486 did not affect the differentiation of T cell but when administered together with relaxin-2, its presence completely abrogated the relaxin-induced Treg increase. #, p < 0.05 vs. control; Kruskal-Wallis ANOVA on ranks for global testing with posthoc Mann-Whitney U- tests for pairwise comparisons (Bonferroni-Holm adjustment of p).
[0081] These findings, therefore, substantiate a stimulatory effect of synthetic human relaxin-2 on the differentiation of peripheral (spleen) regulatory T cells (Treg) in mice and show that these effects are GR-dependent because they could be nullified by a GR-antagonist such as RU-486. DISCUSSION AND SYNOPSIS
[0082] Conventionally, the administration of relaxin-2 in clinical trials and studies has been based on the assumption of a direct and immediate relationship between serum levels and relaxin activity, as is known from insulin. However, as shown in Examples 3 and 4, relaxin-
2 can reverse maladaptive remodeling of the aged heart, but the mechanisms of action are still unknown. In this regard, relaxin-2, although structurally an insulin-like hormone of the insulin superfamily, has been shown to suppress atrial fibrillation, inflammation, and fibrosis in aged rats. However, the administration of relaxin-2 in clinical trials and strudies has still been based i on the assumption of a continuous relation of circulating relaxin levels and relaxin activity. The present application shows that, although the mechanisms of action remain to become elucidated, there is no such direct relationship and that relaxin-2 probably acts by specifically activating genes in tissue development. This is a mechanism of action common to steroids and glucocorticoids as well as to the canonical wnt-signaling pathway. The wnt-signalling pathway is known to play a critical role in embryonic and tissue development which can be inhibited by the circulating proteins of the Dickkopf family.
[0083] In addition, glucocorticoids and corticosteroids are mainstays in the treatment of tissue-damaging autoimmune pathologies such as rheumatoid arthritis, and they are immunosuppressants following organ transplantation. An overwhelming amount of literature exists on the glucocorticoid-mediated regulation of the immune system and in particular on the glucocorticoid-mediated regulation of innate immunity and inflammation. The calcium-binding S100 proteins are universal markers for inflammation and of the innate immune system, notably calprotectin (the complex of S100A8 and S100A9) as well as S100A12. Generally, the presence of S100A12 and calprotectin indicate tissue injury, endothelial cell activation, and inflammation-mediated responses. Cell stress and/or inflammation induce the release of S100 proteins to acellular compartments where they bind cell surface receptors such as RAGE, TLR4, CD147, and GPCR. The interactions between the calcium-binding S100 proteins and their receptors activate intracellular signaling pathways such as AP1 and NFKB, which further initiates multiple cellular processes such as cell differentiation, migration, apoptosis, proliferation, and inflammation; activator protein 1 (AP1), extracellular signal-regulated protein kinase (ERK), G-protein-coupled receptor (GPCR,); interleukin 1 (IL-1); interleukin 7 (IL-7), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IKBO), c-Jun
N-terminal kinase (JNK), p38 mitogen-activated protein kinase (P38), the receptor for advanced glycation end products (RAGE), toll-like receptor 4 (TLR4), TNF receptor-associated factor 2 (Traf2). Mammalian cells secrete calprotectin during the inflammatory response. The exact mechanism by which the complex of S100A8/S100A9 is secreted by mammalian cells during inflammation remains unknown. When released to the extracellular space, S100 proteins have activities in the regulation of immune homeostasis, post-traumatic injury, tissue damage, and inflammation. S100 proteins trigger inflammation through interacting with receptors RAGE and TLR4, and there is evidence that calprotectin (S100A8/S100A9) is an endogenous agonist of TLR4. Binding to TLR4 initiates a signaling cascade and regulates inflammation, cell proliferation, and differentiation in an NFKB-dependent manner Apart from
TLR4, RAGE has also been suggested to bind S100 proteins such as S100A7, S100A12, S100A8/A9 (calprotectin), and SWOB. By interacting with RAGE, S100 proteins activate NFKB, inducing the production of pro-inflammatory cytokines leading to the migration of neutrophils, monocytes, and macrophages. Extracellular S100 proteins are therefore involved in the regulation of cell apoptosis, migration of monocytes, macrophages, neutrophils, lymphocytes, myoblasts, epithelial cells, and endothelial cells. Consequently, the levels of S100A8/A9 complex (calprotectin) and S100A12 in extracellular fluids can be used as biomarkers to assess the degree of inflammatory regulation and tissue injury.
[0084] Liver transplantation and cold storage of livers were studied in mice having the same genetic setup. In this syngeneic mouse model, all immunological responses due to surgical trauma, cold storage, and ischemia injury are therefore mediated by the innate immune system whose effects can be dampened by the administration of corticosteroids. In this syngeneic mouse model, the administration of relaxin-2 at cold storage and/or at reperfusion proved cell-protective and markedly improved posttransplant liver function and survival (cf. Kageyama S et al. in Recombinant relaxin protects liver transplants from ischemia damage by hepatocyte glucocorticoid receptor: From bench-to-bedside, Hepatology 2018,
258-273). Although the authors hypothesize a regulatory role of the relaxin-2-GR complex in the inflammatory injury in liver transplants, the impact of relaxin on the promotion of peripheral
Treg cells and the activation and promotion of immune-suppressive Treg cells was not noted.
With the knowledge of the present invention, continuous treatment of the recipient with relaxin-
2 is warranted as the activated peripheral Treg cells can suppress local immune responses and responses activated by any kind of tissue- and endothelial injury. A continuous administration of relaxin-2 is also warranted as relaxin-2 does not lead to gluconeogenesis as shown in Figs. 5k and 5I. [0085] Concerning the anti-cancer activity of glucocorticoids, for example, De Bono et al. 2014 (Clin Cancer Res 2014, 20:1925-1934, US2006018910) disclose treating hormone- refractory prostate cancer patients with a combination of docetaxel, anti-IGF-IR antibodies, and dexamethasone, since the glucocorticoid receptor is upregulated in refractory prostate cancer cells, and therefore should be inhibited to impair the proliferation of these cancer cells (Ruhr et al., 2018. Clin Cancer Res 24: 927-938). It was found that activation of the glucocorticoid receptor leads to the acquisition of quiescence, subserved by cell cycle arrest through p57 and reprogramming of signaling orchestrated via Insulin Receptor Substrate 2 (IRS2)/Forkhead Box 01 (FOXOI). As synthetic relaxin-2 can be used as a substitute for dexamethasone, it may be favorably used in such therapy without incurring the adverse effects of glucocorticoids and their analogs.
[0086] Boehnert MU in Relaxin as an additional protective model of isolate perfused rat liver, Ann N Y Acad Sci 2005, 1041 :434-440 and Bausys A et al. in Custodiol® supplemented with synthetic human relaxin decreases ischemia-reperfusion injury after porcine kidney transplantation, Int J Mol Sci. 2021 , 22, 11417 describe that relaxin in the perfusion solution reduces ischemia-reperfusion injury (IRI) after kidney or liver
1 transplantation, and that relaxin-2 (RLX) upregulates the expressions of mitochondrial superoxide dismutase-2 (SOD2) and nuclear factor kappa B (NFKB), a regulator of innate immunity. The expression of receptor-interacting serine/threonine-protein kinase 1 (RIPK1), which plays a role in apoptosis and necroptosis, was downregulated compared to controls.
Also downregulated was the expression of mixed lineage kinase domain-like protein (MLKL), which plays a role in tumor necrosis factor (TNF)-induced necroptosis, and the number of caspase 3- and MPO-positive cells was also decreased in the grafts after static cold storage in a solution containing relaxin-2. Static cold storage in a cardioplegic solution is the simplest, most convenient, and least expensive method of organ preservation in clinical practice. While relaxin-2 has been described for its antifibrotic, antioxidant, anti-inflammatory, and cytoprotective properties, there was no evidence that relaxin-2 can be used beneficially as a substitute for glucocorticoids without inducing their Cushingoid adverse effects, particularly gluconeogenesis, and its promotion of immune suppressive Treg cells was also not observed, which, however, makes relaxin-2 a broad-spectrum drug fortissue and endothelial injury, rather than a vasodilator additive in a perfusion solution to prevent ischemia-reperfusion injury.
- SYNOPSIS [0087] In a model of HFpEF, ZSF1 -obese, where both male and female rats on high- fat diet develop this specific type of heart failure, synthetic human relaxin-2 significantly improves diastolic dysfunction, the echocardiographic measure that reflects left ventricular stiffening and subsequent left atrial remodelling (enlargement). These beneficial effects may be attributed to relaxin’s pleiotropic actions, including anti-inflammation, anti-fibrosis, up- regulation of anti-oxidant enzymes, protection of the functionality of the nitric oxide - cGMP- protein kinase G axis responsible for myocardial relaxation (titin phosphorylation), stimulation of active myocardial relaxation via phospholamban and SERCA-2a, as previously reviewed the article by Dschietzig TB, Relaxin-2 for heart failure with preserved ejection fraction (HFpEF): Rationale for future clinical trials, Molecular and Cellular Endocrinology 2019, 487: 54-8.
[0088] The present application discloses a novel mode of drug administration, namely subcutaneous injection of a bolus of relaxin-2 at daily intervals, which not only simplifies chronic clinical use of relaxin-2 and thus increases patient compliance, but also points to an unknown mechanism of its physiological actions and roles. The equipotentiality of a daily bolus of relaxin-
2 compared to continuous subcutaneous infusion could be explained by a slow dissociation of bound relaxin-2 from its cognate receptor, RXFP1 (Valkovic AL et al. in Real-time examination of cAMP activity at relaxin family peptide receptors using a BRET-based biosensor, Pharmacological Research and Perspectives 2018, e00422) and a variety of intracellular signaling cascades in response, and/or by the genomic effects via different pathways including cAMP response elements (CRE), the glucocorticoid receptor as described in this application, protein kinase G-mediated effects, as well as by Wnt-1 -mediated myocardial effects (Martin B et al. in Relaxin reverses maladaptive remodeling of the aged heart through Wnt-signaling,
Scientific Reports 2019, 9: 18545). However, these publications cannot suggest a daily relaxin bolus for the treatment of HFpEF, even when genomic effects may be longer-lasting in nature, since a reversal of the cardiac ageing as proven in Figures 4D to F is very multifactorial which escapes the usual explanations.
SEQUENCE LISTING
<110> Relaxera Pharmazeutis che Gesells chaft mbH
<120> RELAXIN MEDICATION
<130> RLX00257 PC
<160> 10
<170> Patentin version 3 . 5
<210> 1
<211> 29
<212> PRT
<213> Homo sapiens
<400> 1
Asp Ser Trp Met Glu Glu Vai lie Lys Leu Cys Gly Arg Glu Leu Vai 1 5 10 15
Arg Ala Gin lie Ala lie Cys Gly Met Ser Thr Trp Ser
20 25
<210> 2
<211> 24
<212> PRT
<213> Homo sapiens
<400> 2
Glx Leu Tyr Ser Ala Leu Ala Asn Lys Cys Cys His Vai Gly Cys Thr 1 5 10 15
Lys Arg Ser Leu Ala Arg Phe Cys 20
<210> 3
<211> 27
<212> PRT
<213> Homo sapiens
<400> 3
Arg Ala Ala Pro Tyr Gly Vai Arg Leu Cys Gly Arg Glu Phe lie Arg 1 5 10 15
Ala Vai lie Phe Thr Cys Gly Gly Gly Arg Trp
20 25
<210> 4
<211> 24
<212> PRT
<213> Homo sapiens
<400> 4
Asp Vai Leu Ala Gly Leu Ser Ser Ser Cys Cys Lys Trp Gly Cys Ser
1 5 10 15 Lys Ser Glu lie Ser Ser Leu Cys
20
<210> 5
<211> 31
<212> PRT
<213> homo sapiens
<400> 5
Pro Thr Pro Glu Met Arg Glu Lys Leu Cys Gly His His Phe Vai Arg 1 5 10 15
Ala Leu Vai Arg Vai Asp Gly Gly Pro Arg Trp Ser Thr Glu Ala 20 25 30
<210> 6
<211> 26
<212> PRT
<213> Homo sapiens
<400> 6
Ala Ala Ala Thr Asn Pro Ala Arg Tyr Cys Cys Leu Ser Gly Cys Thr 1 5 10 15
Gin Gin Asp Leu Leu Thr Leu Cys Pro Tyr
20 25
<210> 7
<211> 24
<212> PRT
<213> Homo sapiens
<400> 7
Lys Glu Ser Vai Arg Leu Cys Gly Leu Glu Tyr lie Arg Thr Vai lie 1 5 10 15
Tyr lie Cys Ala Ser Ser Arg Trp
20
<210> 8
<211> 21
<212> PRT
<213> Homo sapiens
<400> 8
Glx Asp Leu Gin Thr Leu Cys Cys Thr Asp Gly Cys Ser Met Thr Asp 1 5 10 15
Leu Ser Ala Leu Cys
20
<210> 9
<211> 30 <212> PRT
<213> Homo sapiens
<400> 9
Phe Vai Asn Gin His Leu Cys Gly Ser His Leu Vai Glu Ala Leu Tyr 1 5 10 15
Leu Vai Cys Gly Glu Arg Gly Phe Phe Tyr Thr Pro Lys Thr 20 25 30
<210> 10
<211> 21
<212> PRT
<213> Homo sapiens
<400> 10
Gly lie Vai Glu Gin Cys Cys Thr Ser lie Cys Ser Leu Tyr Gin Leu 1 5 10 15
Glu Asn Tyr Cys Asn
20

Claims

1. A medicament for daily (q.d.) or twice daily (b.i.d.) subcutaneous injection over a prolonged period comprising as bolus a solution of synthetic human relaxin-2 from 75 pg to 2250 micrograms and a pharmacologically acceptable adjuvant, excipient, or diluent.
2. The medicament of claim 1 , wherein synthetic human relaxin-2 is dissolved in 0.5 to 1 .5 ml aqua bidest.
3. The medicament of claim 1 , wherein synthetic human relaxin-2 is dissolved in 0.5 to 1 .5 ml aqueous sodium acetate pH 5.0.
4. The medicament of any preceding claim, wherein the bolus comprises a complex of zinc and human relaxin-2.
5. The medicament of any preceding claim, wherein the bolus comprises a hexamer complex composed of Zn2+ ions and human relaxin-2.
6. The medicament of any preceding claim, wherein the bolus comprises as adjuvant a carbohydrate, preferably selected from mannitol, dextrin, B-dextrin, to adapt osmolality and solubility.
7. The medicament of any preceding claim, wherein the bolus is contained in an injection pen designed for ready-to-use subcutaneous injection.
8. The medicament of any preceding claim, which is contained in a set of pens intended for daily (q.d.) or twice daily (b.i.d.) subcutaneous injections for a period of at least one week, preferably three to twelve months.
9. The medicament of any preceding claim wherein the set of pens is intended for treating heart failure with preserved ejection fraction for a period of at least three months.
10. The medicament of any preceding claim wherein the set of pens is intended for the treatment of a patient suffering from heart failure with preserved ejection fraction for a period of six months or more. 38
11. The medicament of any preceding claim wherein the set of pens is intended for the treatment of a patient suffering from diabetes and heart failure with preserved injection fraction.
12. The medicament of any preceding claim wherein the set of pens is intended for the treatment of a patient suffering from atrial fibrillation and heart failure with preserved injection fraction.
13. The medicament of any preceding claim wherein the set of pens is intended for the treatment of a patient suffering from ischemic heart disease.
14. The medicament of any preceding claim wherein the set of pens is intended for adjunctive medical therapy to coronary angioplasty.
15. The medicament of any preceding claim intended for the protective effects of synthetic human relaxin-2 associated with ischemic heart disease, including the reduction of inflammatory leukocyte and platelet responses, inhibition of the release of proinflammatory and arrhythmogenic mediators by leukocytes and mast cells and other harmful substances generated locally at reperfusion by inflammation, oxidative stress, and necrosis.
16. The medicament of any preceding claim intended for treating a patient requiring a chronic dampening of the innate immune system, of inflammatory responses triggered by the innate immune system and/or suppression through a ligand-activated glucocorticoid receptor.
17. The medicament of any preceding claim intended for the treatment of a patient suspected of having inflammatory responses triggered by the innate immune system and having the following clinical criteria: pre-diabetes (HbA1C > 5.7 and < 6.5 %), obesity (BMI > 30 kg/m2), hypertension (stage 1 or higher according to the 2017 ACC/AHA Guidelines).
18. The medicament of any preceding claim wherein the set of pens is intended for the treatment of a patient who has received an allotransplant and needs a chronic dampening of the innate immune system and inflammatory responses while preventing compromised wound healing, manifestation, or deregulation of diabetes or symptoms of a Cushing’s syndrome.
19. The medicament of any preceding claim intended for the treatment of a transplant patient when one or more of the following four criteria are fulfilled: serum HMGB1 (high- mobility group box protein) greater or equal to 2 ng/ml, serum sTLR4 (soluble Toll-like receptor-4) greater or equal to 0.25 ng/ml, serum sRAGE (soluble receptor of advanced glycation end-products) greater or equal to 0.5 ng/ml, and/or serum calprotectin greater or equal to 4 micrograms/ml.
20. The medicament of any preceding claim intended for the treatment of a patient in need of hormone-refractory cancer therapy, including, but not limited to prostate cancer, breast cancer, or primary cancer therapy through a ligand-activated GR, including, but not limited to Multiple Myeloma, Hodgkin’s Disease, and other Lymphoid Cancers; Kaposi Sarcoma, the synthetic human relaxin-2 being used as supplement and substitute of the glucocorticoid receptor activating hormone.
21 . The medicament of any preceding claim intended for treating a patient suffering from forms of autoimmune or rheumatic disease; ankylosing spondylitis (AS) and spondylarthritis, fibromyalgia, gout, infectious arthritis, lupus, systemic autoimmune disease, osteoarthritis (OA), psoriatic arthritis (PsA) and inflammatory types of arthritis, rheumatoid arthritis (RA).
22. The medicament of any preceding claim intended for treating a patient suffering from SIRS (systemic inflammatory response syndrome), autoimmune or rheumatic diseases, thyroiditis, gastritis, insulitis, sialoadenitis, adrenalitis, oophoritis, glomerulonephritis, polyarthritis, ankylosing spondylitis (AS) and spondylarthritis, fibromyalgia, gout, infectious arthritis, lupus, systemic autoimmune disease, osteoarthritis (OA), psoriatic arthritis (PsA) and inflammatory types of arthritis, rheumatoid arthritis (RA), SARS-Covid 19 und SARS.
* * * * *
PCT/EP2023/0827632023-09-152023-11-22Relaxin medicationPendingWO2025056188A1 (en)

Applications Claiming Priority (2)

Application NumberPriority DateFiling DateTitle
DE102023124949.52023-09-15
DE1020231249492023-09-15

Publications (1)

Publication NumberPublication Date
WO2025056188A1true WO2025056188A1 (en)2025-03-20

Family

ID=88978478

Family Applications (1)

Application NumberTitlePriority DateFiling Date
PCT/EP2023/082763PendingWO2025056188A1 (en)2023-09-152023-11-22Relaxin medication

Country Status (1)

CountryLink
WO (1)WO2025056188A1 (en)

Citations (24)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
CN1035619A (en)*1988-02-261989-09-20基因技术公司Human relaxin formulation
WO1993003755A2 (en)1991-08-191993-03-04Genentech, Inc.Use of relaxin in cardiovascular therapy and in the treatment of neurodegenerative diseases
US5612051A (en)*1995-11-171997-03-18Yue; Samuel K.Method of treating involuntary muscle dysfunction with relaxin hormone
EP0793505A1 (en)1993-10-031997-09-10Knowhow Licensing &amp; Knowhow Transfer B.V.Medicaments comprising relaxin and their use
EP0991947A1 (en)1997-06-232000-04-12The Regents Of The University Of CaliforniaRelaxin levels correlated to ivf/et pregnancy success
WO2000048618A1 (en)1999-02-182000-08-24Yue Samuel KMethod of preventing and treating symptoms of aging and neurodegenerative dysfunctions with relaxin
WO2001058468A1 (en)2000-02-092001-08-16Connetics CorporationUse of relaxin to treat diseases related to vasoconstriction
WO2002040500A2 (en)2000-11-162002-05-23Immundiagnostik AgMethod of prognosis and diagnosis of congestive heart failure
EP1473034A1 (en)2003-03-172004-11-03Lagow GmbHUse of an inhibitor of the phosphodiesterase-4-enzyme (PDE4) alone or in combination with relaxin and derivatives thereof for the manufacture of a medicament to promote decidualization of endometrial cells and to increase fertility
US20060018910A1 (en)2004-07-162006-01-26Pfizer IncCombination treatment for non-hematologic malignancies
DE102005040492A1 (en)2004-08-272006-03-16Immundiagnostik AgPerfusion- and preservation solution, useful for transplants and for perfusing and preserving organs for transplantation, comprises relaxin
EP1696948A1 (en)2003-12-242006-09-06Bigazzi, MarioUse of relaxin as adjuvant in the differentiation of stem cells for the reconstruction of tissues
EP1765149A2 (en)2004-03-192007-03-28Kirk P. ConradUse of relaxin to increase arterial compliance
WO2007115414A1 (en)2006-04-112007-10-18University Of GuelphModified h2 relaxin for tumor suppression
EP1854476A2 (en)2000-02-092007-11-14Bas Medical, Inc.Use of relaxin to treat diseases related to vasoconstriction
EP1909809A2 (en)2005-06-132008-04-16Sky Biohealth Solutions, Inc.Method and compositions for the treatment of diabetes and related complications
US7878978B2 (en)2004-03-182011-02-01University Of Pittsburgh- Of The Commonwealth System Of Higher EducationUse of relaxin to increase arterial compliance
WO2014059174A1 (en)2012-10-102014-04-17University Of Pittsburgh - Of The Commonwealth System Of Higher EducationUse of relaxin to treat atrial fibrillation
EP2723366A1 (en)2011-06-242014-04-30Bold Venture LLCRelaxin proteins for use in the diagnosis, prevention or treatment of multiple sclerosis
EP2817026A1 (en)2012-02-222014-12-31Immundiagnostik AGRelaxin for treating patients afflicted of impaired glucose tolerance
EP2829280A1 (en)2008-05-162015-01-28Corthera, Inc.Treating dyspnea associated with acute heart failure with H2 relaxin
WO2017042577A1 (en)2015-09-102017-03-16Bv Biomed LtdNovel therapy
EP3145534A1 (en)2014-05-232017-03-29Immundiagnostik AGMethod and composition for treating heart failure with preserved ejection fraction
WO2022037469A1 (en)2020-08-172022-02-24成都奥达生物科技有限公司Long-acting relaxin-2 analog

Patent Citations (35)

* Cited by examiner, † Cited by third party
Publication numberPriority datePublication dateAssigneeTitle
CN1035619A (en)*1988-02-261989-09-20基因技术公司Human relaxin formulation
WO1993003755A2 (en)1991-08-191993-03-04Genentech, Inc.Use of relaxin in cardiovascular therapy and in the treatment of neurodegenerative diseases
EP0793505A1 (en)1993-10-031997-09-10Knowhow Licensing &amp; Knowhow Transfer B.V.Medicaments comprising relaxin and their use
US5612051A (en)*1995-11-171997-03-18Yue; Samuel K.Method of treating involuntary muscle dysfunction with relaxin hormone
EP0991947A1 (en)1997-06-232000-04-12The Regents Of The University Of CaliforniaRelaxin levels correlated to ivf/et pregnancy success
WO2000048618A1 (en)1999-02-182000-08-24Yue Samuel KMethod of preventing and treating symptoms of aging and neurodegenerative dysfunctions with relaxin
EP1253929B1 (en)2000-02-092007-05-23BAS Medical, Inc.Use of relaxin to treat diseases related to vasoconstriction
WO2001058468A1 (en)2000-02-092001-08-16Connetics CorporationUse of relaxin to treat diseases related to vasoconstriction
US6723702B2 (en)2000-02-092004-04-20Ras Medical, Inc.Use of relaxin treat diseases related to vasoconstriction
US20020019349A1 (en)2000-02-092002-02-14Conrad Kirk P.Use of relaxin treat diseases related to vasoconstriction
EP1854476A2 (en)2000-02-092007-11-14Bas Medical, Inc.Use of relaxin to treat diseases related to vasoconstriction
WO2002040500A2 (en)2000-11-162002-05-23Immundiagnostik AgMethod of prognosis and diagnosis of congestive heart failure
EP1473034A1 (en)2003-03-172004-11-03Lagow GmbHUse of an inhibitor of the phosphodiesterase-4-enzyme (PDE4) alone or in combination with relaxin and derivatives thereof for the manufacture of a medicament to promote decidualization of endometrial cells and to increase fertility
EP1696948A1 (en)2003-12-242006-09-06Bigazzi, MarioUse of relaxin as adjuvant in the differentiation of stem cells for the reconstruction of tissues
US20140057832A1 (en)2004-03-182014-02-27University Of Pittsburgh - Of The Commonwealth System Of Higher EducationUse of relaxin to increase arterial compliance
US7878978B2 (en)2004-03-182011-02-01University Of Pittsburgh- Of The Commonwealth System Of Higher EducationUse of relaxin to increase arterial compliance
US8602998B2 (en)2004-03-182013-12-10University of Pittsburgh—of the Commonwealth System of Higher EducationUse of relaxin to increase arterial compliance
EP1765149A2 (en)2004-03-192007-03-28Kirk P. ConradUse of relaxin to increase arterial compliance
US20060018910A1 (en)2004-07-162006-01-26Pfizer IncCombination treatment for non-hematologic malignancies
DE102005040492A1 (en)2004-08-272006-03-16Immundiagnostik AgPerfusion- and preservation solution, useful for transplants and for perfusing and preserving organs for transplantation, comprises relaxin
EP1909809A2 (en)2005-06-132008-04-16Sky Biohealth Solutions, Inc.Method and compositions for the treatment of diabetes and related complications
WO2007115414A1 (en)2006-04-112007-10-18University Of GuelphModified h2 relaxin for tumor suppression
EP2829280A1 (en)2008-05-162015-01-28Corthera, Inc.Treating dyspnea associated with acute heart failure with H2 relaxin
EP2723366A1 (en)2011-06-242014-04-30Bold Venture LLCRelaxin proteins for use in the diagnosis, prevention or treatment of multiple sclerosis
EP2817026A1 (en)2012-02-222014-12-31Immundiagnostik AGRelaxin for treating patients afflicted of impaired glucose tolerance
US20170342120A1 (en)2012-10-102017-11-30University Of Pittsburgh - Of The Commonwealth System Of Higher EducationUse of relaxin to treat atrial fibrillation
US20150232526A1 (en)2012-10-102015-08-20University Of Pittsburgh-Of The Commonwealth System Of Higher EducationUse of relaxin to treat atrial fibrillation
US20200087371A1 (en)2012-10-102020-03-19University Of Pittsburgh - Of The Commonwealth System Of Higher EducationUse of relaxin to treat atrial fibrillation
WO2014059174A1 (en)2012-10-102014-04-17University Of Pittsburgh - Of The Commonwealth System Of Higher EducationUse of relaxin to treat atrial fibrillation
EP3145534A1 (en)2014-05-232017-03-29Immundiagnostik AGMethod and composition for treating heart failure with preserved ejection fraction
US10226512B2 (en)2014-05-232019-03-12Relaxera Pharmazeutische Gesellschaft mbH & Co. KGMethod for treating heart failure with preserved ejection fraction by administering human relaxin-2
EP3347037A1 (en)2015-09-102018-07-18BvBiomed Ltd.Novel therapy
WO2017042577A1 (en)2015-09-102017-03-16Bv Biomed LtdNovel therapy
EP3347037B1 (en)2015-09-102020-12-16BVBiomedical LimitedNovel therapy
WO2022037469A1 (en)2020-08-172022-02-24成都奥达生物科技有限公司Long-acting relaxin-2 analog

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
ALEXIOU K ET AL.: "Relaxin is a candidate drug for lung preservation: relaxin induced protection of rat lungs from ischemia-reperfusion injury", J HEART LUNG TRANSPLANT, vol. 29, 2010, pages 454 - 460, XP026981546
ANN N Y, ACAD SCI, vol. 1041, 2005, pages 434 - 440
BANI D: "Recombinant human H2 relaxin (serelaxin) as a cardiovascular drug: aiming at the right target", DRUG DISCOVERY TODAY, vol. 25, no. 7, 2020, pages 1239 - 44, XP086225629, DOI: 10.1016/j.drudis.2020.04.014
BAUSYS A ET AL.: "Custodiol@ supplemented with synthetic human relaxin decreases ischemia-reperfusion injury after porcine kidney transplantation", INT J MOL SCI, vol. 22, 2021, pages 11417
COLLINO M ET AL.: "Acute treatment with relaxin protects the kidney against ischemia-reperfusion injury", J CELL MOL MED, vol. 17, 2013, pages 1494 - 1505
COULSON CC ET AL.: "Central hemodynamic effects of recombinant human relaxin in the isolated, perfused rat heart model", OBSTETRICS & GYNECOLOGY, vol. 87, no. 4, 1996, pages 610 - 612
DE BONO ET AL., CLIN CANCER RES, vol. 20, 2014, pages 1925 - 1934
DILASCIO G ET AL.: "Cellular retrograde cardiomyoplasty and relaxin therapy for postischemic myocardial repair in a rat model", TEX HEART INST J, vol. 39, 2012, pages 488 - 499
DSCHIETZIG T ET AL.: "Autoregulation of human relaxin-2 gene expression critically involves relaxin and glucocorticoid receptor binding to glucocorticoid response half-sites in the relaxin-2 promoter", REGUL PEPT, vol. 155, 2009, pages 163 - 73, XP026158590, DOI: 10.1016/j.regpep.2009.03.001
DSCHIETZIG T ET AL.: "Plasma levels and myocardial expression of relaxin-2 are increased in human heart failure", CIRCULATION, vol. 102, no. 18, 2000, pages 594
DSCHIETZIG T ET AL.: "Relaxin - a pleiotropic hormone and its emerging role for experimental and clinical therapeutics", PHARMACOL THER, vol. 112, 2006, pages 38e56
DSCHIETZIG T ET AL.: "Relaxin: a pregnancy hormone as a central player of body fluid and circulation homeostasis", CMLS, vol. 60, 2003, pages 688 - 700, XP008145479, DOI: 10.1007/s00018-003-2169-x
DSCHIETZIG T ET AL.: "RXFP1-inactive relaxin activates human glucocorticoid receptor: further investigations into the relaxin-GR pathway", REGUL PEPT, vol. 154, 2009, pages 77 - 84, XP026079176, DOI: 10.1016/j.regpep.2008.11.010
DSCHIETZIG T ET AL.: "The pregnancy hormone relaxin binds to and activates the human glucocorticoid receptor", ANN N Y ACAD SCI, vol. 1041, 2005, pages 256 - 71, XP071401576, DOI: 10.1196/annals.1282.039
DSCHIETZIG TB ET AL.: "Identification of the pregnancy hormone relaxin as a glucocorticoid receptor agonist", FASEB J, vol. 18, 2004, pages 1536 - 1538
DSCHIETZIG TB: "Relaxin-2 for heart failure with preserved ejection fraction (HFpEF): Rationale for future clinical trials", MOLECULAR AND CELLULAR ENDOCRINOLOGY, vol. 487, 2019, pages 54 - 8, XP085650819, DOI: 10.1016/j.mce.2019.01.013
HALLS M ET AL.: "Recent advances in the understanding of the pharmacology and biological roles of relaxin family peptide receptors 1-4, the receptors for relaxin family peptides", PHARM REV, vol. 67, no. 2, 2015, pages 389 - 440
HAMDANI N ET AL.: "Myocardial Titin Hypophosporylation importantly", CIRCULATION HEART FAILURE, vol. 6, no. 6, 2013, pages 1239 - 1249
HEMMERLING M ET AL.: "Selective Nonsteroidal Glucocorticoid Receptor Modulators for the Inhaled Treatment of Pulmonary Diseases", J. MED. CHEM., vol. 60, no. 20, 2017, pages 8591 - 8605, XP055825954, DOI: 10.1021/acs.jmedchem.7b01215
HISAW FL: "Experimental relaxation of the pubic ligament of the guinea pig", PROC. SOC. EXP. BIOL. MED, vol. 23, 1926, pages 661 - 663
JAKUBAUSKIENE L ET AL.: "Relaxin positively influences ischemia-reperfusion injury in solid organ transplantation: a comprehensive review", INT J MOL SCI, vol. 21, no. 2, 2020, pages 631
KAGEYAMA S ET AL., RELAXIN IN LIVER TRANSPLANTATION: A PERSONAL PERSPECTIVE, MOL CELL ENDOCRINOL, vol. 487, 2019, pages 75 - 79
KAGEYAMA S ET AL.: "Recombinant relaxin protects liver transplants from ischemia damage by hepatocyte glucocorticoid receptor.- From bench-to-bedside", HEPATOLOGY, 2018, pages 258 - 273
KHANNA D ET AL.: "A randomized, double-Blind, placebo-controlled trial of recombinant human relaxin in the treatment of systemic sclerosis with diffuse scleroderma", ARTHRITIS RHEUM, vol. 60, no. 4, 2009, pages 1102 - 1111
MARGARITA DOMINQUEZ-VALLARDAVID A. HAFLER: "Regulatory Tcells in autoimmune disease", NATURE IMMUNOLOGY, vol. 19, 2018, pages 665 - 673
MARTIN B ET AL.: "Relaxin reverses maladaptive remodeling of the aged heart through VVnt-signaling", SCIENTIFIC REPORTS, vol. 9, 2019, pages 18545
MASINI E. ET AL.: "Relaxin counteracts myocardial damage induced by ischemia-reperfusion in isolated guinea pig hearts: evidence for an involvement of nitric oxide", ENDOCRINOLOGY, vol. 138, 1997, pages 4713 - 4720
METRA M, TEERLINK ET AL.: "Effects of serelaxin in patients with acute heart failure", N ENGL J MED, vol. 381, 2019, pages 716 - 726
MUPPIDI, A ET AL.: "Design and Synthesis of Potent, Long-Acting Lipidated Relaxin-2 Analogs", BIOCONJUGATE CHEM, vol. 30, 13 December 2018 (2018-12-13), pages 83 - 89, XP055838187, DOI: 10.1021/acs.bioconjchem.8b00764
NAIR, V.B ET AL.: "Synthetic Covalently Linked Dimeric Form of H2 Relaxin Retains Native RXFPIActivity and has improved in Vitro Serum Stability", BIOMED RESEARCH INT, 31 December 2015 (2015-12-31)
NICOLAIDES N ET AL.: "Glucocorticoid Receptor", 2021
RAZEGHI P ET AL.: "Downregulation of metabolic gene expression in failing human heart before and after mechanical unloading", CARDIOLOGY, vol. 97, no. 4, 2002, pages 203 - 9
RUHR ET AL., CLIN CANCER RES, vol. 24, 2018, pages 927 - 938
SCHAUER A ET AL.: "ZSF1 rat as animal model for HFpEF: Development of reduced diastolic function and skeletal muscle dysfunction", ESC HEART FAILURE, vol. 7, 2020, pages 2123 - 2134
SCHIATTARELLA G.G ET AL.: "Nitrosative stress drives heart failure with preserved ejection fraction", NATURE, vol. 568, 2019, pages 351 - 356, XP036900417, DOI: 10.1038/s41586-019-1100-z
SEIBOLD JR ET AL.: "Recombinant human relaxin in the treatment of scleroderma. A randomized, double-blind, placebo-controlled trial", ANN INTERN MED, vol. 132, no. 11, 2000, pages 871 - 9
SEIBOLD JR ET AL.: "Safety and pharmacokinetics of recombinant human relaxin in systemic sclerosis", J RHEUMATOL, vol. 25, no. 2, 1998, pages 302 - 7, XP009066594
TAMAKI N ET AL., AM J PHYSIOL GASTROINTEST LIVER PHYSIOL, vol. 294, 2008, pages G499
TEERLINK JR ET AL.: "Serelaxin, recombinant human relaxin-2, for treatment of acute heart failure (RELAX AHF): a randomised, placebo-controlled trial", LANCET, 2012, Retrieved from the Internet <URL:http://dx.doi.org/10.1016/S0140-6736(12)61855-8>
TEICHMANN SL ET AL.: "Relaxin: a review of the biology and potential role in treating heart failure", CURR HEART FAIL REP, vol. 7, 2010, pages 75 - 82
VALKOVICAL ET AL.: "Real-time examination of cAMP activity at relaxin family peptide receptors using a BRET-based biosensor", PHARMACOLOGICAL RESEARCH AND PERSPECTIVES, 2018, pages e00422
VILLALBA-ORERO M ET AL.: "g ultrasound as a translational approach for non-invasive assessment of heart failure with reduced or preserved ejection fraction in mice", CARDIOVASCULAR RESEARCH, vol. 113, 2017, pages 1113 - 1123

Similar Documents

PublicationPublication DateTitle
JP5261620B2 (en) Alopecia treatment
Wu et al.Ghrelin attenuates sepsis-induced acute lung injury and mortality in rats
Li et al.Berberine improves pressure overload-induced cardiac hypertrophy and dysfunction through enhanced autophagy
Jankowski et al.Anti-inflammatory effect of oxytocin in rat myocardial infarction
US10864229B2 (en)Use of nitrite salts in chronic ischemia
Rao et al.Atrial natriuretic peptide: structure, function, and physiological effects: a narrative review
Samuel et al.Antifibrotic actions of serelaxin–new roles for an old player
US20180325869A1 (en)Targeting of the formyl-peptide receptor 2/lipoxin a4 receptor (fpr2/alx) for treatment of heart disease
ES2648051T3 (en) Use of mono and dicarboxylic acid amides in the treatment of kidney diseases
Xu et al.Ghrelin ameliorates hypoxia-induced pulmonary hypertension via phospho-GSK3 b/b-catenin signaling in neonatal rats
AU2023332056A1 (en)Immunosuppressive medicament and method of treatment
RU2663455C2 (en)COMBINATION OF SOMATOSTATIN ANALOGUES WITH 11β-HYDROXYLASE INHIBITORS
JP6824888B2 (en) Methods and compositions for treating muscle disorders and disorders
CN109475604B (en)Dosing and use of long-acting CLR/RAMP agonists
WO2025056188A1 (en)Relaxin medication
JP2019516778A (en) Prevention and / or treatment of ischemic injury or ischemia reperfusion injury
CN117018202A (en)Novel use of chemokine receptor CCR6 inhibitors for preventing psoriasis recurrence
CarrThe Mechanism of the Prolonged Action of the Single-Chain Insulin, 70-01
KR20190138666A (en) Use of isofumulus and its derivatives for the treatment of polycystic ovary syndrome
JP2019505503A (en) The effect of Lantus on cardiac metabolism
Asensio et al.ZFP-36 proteins in monocyte and macrophage inflammatory signaling and differentiation. Model-ling their post-translational modifications, intracellular locations, interactome, structural hetero-geneity and halflife
WO2025017193A1 (en)Methods of treating cardiomyopathy induced by metabolic inflexibility
JP2025040825A (en) Pharmaceutical composition and method for preventing or treating interstitial pneumonia
JP2019094333A (en)Medicine for the prevention or treatment of heart failure
CN119894515A (en)Methods of treating obesity with luteinizing hormone receptor agonists

Legal Events

DateCodeTitleDescription
121Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number:23814112

Country of ref document:EP

Kind code of ref document:A1


[8]ページ先頭

©2009-2025 Movatter.jp