WO2024245719A1 - Inhibiteurs de sortiline pour le traitement de patients atteints de tumeurs neuroendocrines fonctionnelles - Google Patents
Inhibiteurs de sortiline pour le traitement de patients atteints de tumeurs neuroendocrines fonctionnelles Download PDFInfo
- Publication number
- WO2024245719A1 WO2024245719A1 PCT/EP2024/062984 EP2024062984W WO2024245719A1 WO 2024245719 A1 WO2024245719 A1 WO 2024245719A1 EP 2024062984 W EP2024062984 W EP 2024062984W WO 2024245719 A1 WO2024245719 A1 WO 2024245719A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- sortilin
- inhibitor
- carbamoyl
- benzoic acid
- methylpyridin
- 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
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/44—Non condensed pyridines; Hydrogenated derivatives thereof
- A61K31/445—Non condensed piperidines, e.g. piperocaine
- A61K31/4523—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
- A61K31/4545—Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
Definitions
- the present invention relates to the field of anti-cancer therapies, more specifically to therapies with the aim of improving the health of patients suffering from functional neuroendocrine tumors (NETs) or functional neuroendocrine neoplasms (NENs).
- NETs functional neuroendocrine tumors
- NNNs functional neuroendocrine neoplasms
- the invention discloses the use of sortilin inhibitors for treating a condition associated with the release of an endocrine and/or a paracrine factor such as serotonin by a functional neuroendocrine neoplasm in a subject, e.g., carcinoid syndrome, carcinoid heart disease, and/or carcinoid crisis.
- the sortilin inhibitor can be provided in a pharmaceutical composition and may be a small molecule inhibitor having a molecular weight of 800 daltons or less, an anti-sortilin antibody, an inhibitory nucleic acid, a proteolysis-targeting chimera, a peptide-based inhibitor or a combination thereof.
- NET Neuroendocrine tumors
- NETs are rare tumors arising from hormone-producing cells that have an incidence of about 2-5 per 100,000 population. During the last decades, the age- adjusted incidence rate increased significantly from 1.09 (1973) to 6.98 per 100,000 (2012) (Dasari et al., 2017). The cause of the development of NETs is largely unknown, but in some cases, it can be attributed to genetic alterations in neuroendocrine cells. NETs comprise cells exerting endocrine as well as neural cell characteristics. While they can occur in every organ, they are particularly predominant in the gastroenteropancreatic system (Rindi et al., 2020).
- NETs are classified as “functional”, meaning they are characterized by their ability to produce and release excessive amounts of bioactive substances, especially hormones and biogenic amines such as serotonin. Once these substances are distributed throughout the body via the blood circulation, affected patients frequently develop various hormone-induced symptoms and syndromes that further limit their well-being and overall health in addition to the immediate effects caused by the tumor itself.
- One of the most common hormone mediated syndromes that occur secondary to neuroendocrine tumors is known as carcinoid syndrome. Carcinoid syndrome occurs mainly due to an over-secretion of serotonin, which can be observed in 32% of all small intestinal NETs (Halperin et al., 2017).
- SSA somatostatin analogues
- PRRT Peptide directed radiotherapy
- the present invention provides a sortilin inhibitor for use in treating a condition associated with the release of an endocrine and/or a paracrine factor by a functional neuroendocrine tumor in a subject.
- Also disclosed herein is a method of treating a condition associated with the release of an endocrine and/or a paracrine factor by a functional neuroendocrine tumor in a subject, wherein the method comprises a step of administering to the subject a therapeutically effective amount of a sortilin inhibitor.
- Sortilin also known as neurotensin receptor 3 or SORT1 , is a member of the VPS10P domain sorting receptor family, a group of transmembrane receptors which are involved in sorting and intracellular trafficking of a broad range of ligands (Malik et al., 2020). Sortilin furthermore serves as a co-receptor for the precursor of nerve growth factor (pro-NGF) and the 75-kDa neurotrophin receptor (p75NTR). It has been shown that sortilin is expressed in many healthy as well as in many cancer cells like breast cancer or lung cancer (Roselli et al., 2015: Al- Akhrass et al., 2017; Gao et al., 2018). Expression was also found in NETs and was attributed to cell migration and adhesion (Kim et al., 2018). However, only a fraction of the stained tissue samples were positive for sortilin expression (Kim et al., 2018).
- NETs and their cells are characterized by the cell type-specific presence of so-called neurosecretory vesicles.
- Synaptophysin has been shown in the past to be an essential molecular component of these neurosecretory vesicles and can be used as a central marker molecule for the immunohistochemical diagnosis of NETs. Release of the neurosecretory vesicles leads to the dispersal of the endocrine and paracrine factors that cause NET-specific symptoms and syndromes.
- the present inventors thus addressed the molecular mechanisms that control the release of neurosecretory vesicles.
- the inventors discovered sortilin to be a central regulatory molecule of the neurosecretory release of the endocrine and/or paracrine factors.
- the inventors were able to show that inhibition of sortilin using a suitable sortilin inhibitor effectively prevents the release of these factors by the cells of NETs.
- Treating a condition is herein to be understood to comprise a curative medical therapy of a subject with the intent to cure, ameliorate or stabilize a condition.
- the term “condition” refers to a disease, a syndrome, a disorder or a particular physiological state of an organism that is either manifested by distinct symptoms or can be diagnosed by using established markers capable of recognizing said state.
- treating a condition is intended to mean that the progression of the condition is to be slowed, stopped or, preferably, reversed to allow for a perceivable improvement of the subject s well-being and health.
- the sortilin inhibitor may also be for use in preventing a condition associated with the release of an endocrine and/or a paracrine factor by a functional neuroendocrine tumor in a subject.
- Preventing a condition refers to precluding said condition from happening in the first place. Preventing a condition thus also explicitly includes the prophylactic treatment of a subject.
- the subject typically is a mammal, e.g., a human, non-human primate, horse, pig, rabbit, dog, sheep, goat, cow, camel, cat, guinea pig, rat or mouse.
- the subject is a human.
- neuroendocrine neoplasm NNN
- neuroendocrine tumor NET
- NETs originate from specialized cells of the body’s neuroendocrine system that exhibit traits of both hormone-producing endocrine cells and nerve cells.
- NETs most commonly develop in the gastrointestinal tract, as about 40% of all NETs arise in the colon and small intestine and 4% in the appendix. However, many NETs also develop in the lung (approximately 30%) and pancreas (approximately 7%).
- NETs can secrete various endocrine and/or paracrine factors such as, e.g., hormones and other vasoactive substances such as biogenic amines.
- endocrine and/or paracrine factors such as, e.g., hormones and other vasoactive substances such as biogenic amines.
- the ability to secrete endocrine and/or paracrine factors divides NETs in “functional” and “non-functional”. Accordingly, in the context of the present invention, the term
- “functional NET” (often also referred to as “clinically functioning” or “symptomatic” NET) is herein understood to refer to a NET that generates and secretes endocrine and/or paracrine factors, e.g., vasoactive biogenic amines or hormones.
- the functional NET preferably is a gastrointestinal NET (i.e., a colon NET, a small intestine NET or an appendix NET).
- the NET may however also be a lung NET or a pancreas NET.
- NETs may also develop in or on adrenal glands and are referred to as pheochromocytoma and paraganglioma. Therefore, in some embodiments, the NET may also be a pheochromocytoma or a paraganglioma.
- condition associated with the release of an endocrine and/or paracrine factor by a functional neuroendocrine tumor typically refers to a condition that is associated with the NET but is mediated by one or more endocrine and/or paracrine factor, e.g., a hormones and/or vasoactive substance, secreted by said NET.
- An endocrine factor is a signaling molecule of the endocrine system that is released by an internal gland. It exerts its effects far from its site of production, i.e., it is transported via the blood circulation to a different organ, where it can regulate physiology and/or behaviour.
- Endocrine factors are more commonly referred to as hormones. They can comprise various types of substances, including eicosanoids such as. e.g., prostaglandins, steroids, amino acid derivatives as wells as proteins and peptides.
- eicosanoids such as. e.g., prostaglandins, steroids, amino acid derivatives as wells as proteins and peptides.
- paracrine factors In contrast to endocrine factors, paracrine factors are not distributed via the blood circulation to distant targets but diffuse over relatively short distances. Thus, they exert local effects in the environment in which they are produced.
- endocrine and paracrine factors in the context of the invention need not necessarily refer to two different groups of biologically active substances, but may just as well be used to ascribe different (i.e., endocrine and paracrine) functions to one and the same factor.
- the factor has at least endocrine functions.
- the endocrine and/or paracrine factor released by the functional neuroendocrinic tumor and contributing to the condition to be treated preferably is a biogenic amine such as serotonin, histamine or a (poly-)peptide such as bradykinin, tachykinin (substance P), prostaglandin and/or kallikrein, preferably serotonin.
- a biogenic amine such as serotonin, histamine or a (poly-)peptide such as bradykinin, tachykinin (substance P), prostaglandin and/or kallikrein, preferably serotonin.
- the endocrine and/or paracrine factor secreted by the NET may also be, e.g., adrenaline, gastrin, insulin, glucagon, somatostatin, corticotropin-releasing hormone, adrenocorticotropin, somatoliberin, somatotropin, parathyroid hormone-related protein, calcitonin, calcitonin gene-related peptide, vasoactive intestinal polypeptide, chromogranin A, katacalcin, neuropeptide Y, bombesin/gastrin releasing peptide (GRP), alpha-human chorionic gonadotropin (aHCG). thyroid stimulating hormone-like peptide, cholecystokinin, adrenomedullin, vascular endothelial growth factor (VEGF) and any combination thereof.
- adrenaline gastrin
- insulin glucagon
- somatostatin corticotropin-releasing hormone
- adrenocorticotropin a
- the condition associated with the release of an endocrine and/or paracrine factor by a functional neuroendocrine tumor is carcinoid syndrome.
- Carcinoid syndrome occurs in about 10% of all neuroendocrine tumors and is characterized by various symptoms, including skin flushing, in particular at the head and the upper part of the thorax, facial skin lesions, diarrhea, oftentimes associated with abdominal cramping and pain, bronchoconstriction as well as a rapid heartbeat. Further symptoms include malabsorption, fatigue, muscle loss and cognitive impairment.
- the liver is capable of neutralizing aberrantly secreted hormones and other biologically active substance before they have a chance to travel through the body and cause the symptoms associated with carcinoid syndrome.
- carcinoid syndrome may also arise in the absence of liver metastases due to neuroendocrine tumors in the bronchi, as their vasoactive products may reach the blood circulation before passing through the liver.
- Many of the symptoms associated with carcinoid syndrome primarily develop due to an altered tryptophan metabolism in the neuroendocrine tumor leading to the release of serotonin into the blood stream.
- additional vasoactive substances are thought to contribute to the manifestations of carcinoid syndrome, including histamine, tachykinins, kallikrein and prostaglandins.
- the condition associated with the release of endocrine and/or paracrine factors by a functional neuroendocrine neoplasm may also be carcinoid crisis.
- Carcinoid crisis refers to an extreme exacerbation of carcinoid syndrome due to prolonged and excessive release of endocrine and/or paracrine factors, in particular serotonin, by the NET.
- Carcinoid crisis manifests itself by symptoms including profound flushing, widely and rapidly fluctuating blood pressure, arrythmia, tachycardia, and bronchospasm. It may also occur in patients in which no carcinoid syndrome has been diagnosed before.
- the condition associated with the release of an endocrine and/or paracrine factor by a functional neuroendocrine neoplasm may be carcinoid heart disease.
- Carcinoid heart disease is the result of valvular damage related to the vasoactive factors released by the neuroendocrine tumor affecting, in particular, the tricuspid and the pulmonic valves. In many instances tissue injury around the heart valves is followed by plaque and fibrosis formation. Carcinoid heart disease typically develops as a consequence of prolonged carcinoid syndrome.
- condition associated with the release of an endocrine and/or paracrine factor by a functional neuroendocrine tumor may also refer to merely one of the herein described symptoms of carcinoid syndrome, carcinoid crisis or carcinoid heart disease.
- the condition may be any symptom selected from skin flushing, facial skin lesions, diarrhea, abdominal cramping and pain, bronchoconstriction, rapid heartbeat, malabsorption, fatigue, muscle loss, cognitive impairment, widely and rapidly fluctuating blood pressure, arrythmia, tachycardia, bronchospasm as well as tissue injury around the heart valves followed by plaque and fibrosis formation.
- the excessive secretion of serotonin by NETs can further cause a depletion of tryptophan leading to niacin deficiency, and thus pellagra in the patient, which increases the risk of developing dermatitis, dementia, and diarrhea.
- NETs Many other hormones can be secreted by NETs, most commonly growth hormone (somatotropin) that can cause acromegaly, or cortisol, that can cause Cushing's syndrome. Excessive secretion of insulin results in low blood sugar levels, whereas uncontrolled release of glucagon is associated with the development of skin rashes and a disturbed sugar metabolism. Release of vasoactive intestinal polypeptide may cause diarrhea and increased urination (Verner-Morrison syndrome) and excessive secretion of gastrin increases the acidity in the stomach and small intestine and thus may cause heartburn, diarrhea and gastrointestinal ulcers (Zollinger-Ellison syndrome).
- condition associated with the release of an endocrine and/or paracrine factor by a functional neuroendocrine neoplasm may thus also be niacin deficiency, pellagra, dermatitis, dementia, acromegaly, Cushing's syndrome, low blood sugar, skin rashes, disturbed sugar metabolism, Verner-Morrison syndrome or Zollinger-Ellison syndrome.
- Inhibitors may thus also be niacin deficiency, pellagra, dermatitis, dementia, acromegaly, Cushing's syndrome, low blood sugar, skin rashes, disturbed sugar metabolism, Verner-Morrison syndrome or Zollinger-Ellison syndrome.
- an inhibitor is understood as any compound, substance or composition capable of inhibiting a biological process or a chemical reaction.
- sortilin inhibitor refers to any compound, substance or composition (such as an antibody, protein, nucleic acid or other molecule) that interferes with, blocks, or otherwise attenuates either the expression and/or stability of sortilin itself or the interaction of sortilin with at least one of its ligands, e.g., with pro-neurotrophin.
- the sortilin inhibitor may for instance be a sortilin antagonist that competes with a naturally occurring sortilin ligand for binding to sortilin.
- the sortilin inhibitor of the present invention may prevent the protein- protein interaction between a sortilin protein and its ligand pro-neurotrophin, thereby preventing the formation of the protein complex usually formed between sortilin, pro- neurotrophin and the p75NTR receptor, or resulting in the formation of a low affinity complex, which is biologically less active or inactive or has minimal activity.
- the sortilin inhibitor may completely prevent the interaction between sortilin and its ligands, or may do so only partially, in which case the complex between sortilin and its ligands may still form but may have reduced biological potency.
- the sortilin inhibitor may also interfere with, block or otherwise attenuate sortilin function.
- the sortilin inhibitor may be a small molecule drug having a molecular weight of 800 daltons or less, an anti-sortilin antibody, an inhibitory nucleic acid or a peptide-based inhibitor.
- the sortilin inhibitor is a small molecule drug.
- small molecule 7 refers to an organic compound having a molecular weight of less than 800 Da. The low molecular weight of small molecule drugs enables rapid diffusion of the compounds across cell membranes and increases their oral bioavailability.
- the small molecule inhibitor of sortilin is 2-[[(6-methyl-2-pyridinyl)amino]carbonyl]- 5-(trifluoromethyl)-benzoic acid, which is better known under the name AF38469.
- AF38469 was first described by Schroder et al., in 2014 and is orally bioavailable and highly selective for sortilin over several of its naturally occurring ligands, including, e.g., neurotensin-1. As demonstrated in the Examples below, the inventors successfully used AF38469 as a sortilin inhibitor to suppress secretion of serotonin by NEN cells.
- the small molecule sortilin inhibitor may be (2S)-2- ⁇ [(7-hydroxy-4-methyl-2-oxo- 2H-chromen-8-yl)methyl]amino ⁇ -4-methylpentanoic acid (commonly referred to as AF40431 ), which was the first sortilin inhibitor ever identified, but exhibits comparably poor solubility and membrane permeability.
- the small molecule inhibitor of sortilin may also be 1-[2-(2-tert-butyl- 5-methylphenoxy)-ethyl]-3-methylpiperidine (MPEP). as previously described in Lee et aL, 2014. Unlike e.g., AF38469, which interferes with the interactions of sortilin with its ligands,
- MPEP appears to inhibit sortilin by increasing its degradation, thereby suppressing sortilin protein levels.
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2021/186054 A1 , which is hereby incorporated in its entirety by reference.
- the sortilin inhibitor may thus be a sortilin antagonist of Formula I: or a pharmaceutically acceptable salt, solvate, hydrate, geometrical isomer, tautomer, optical isomer, N-oxide, and/or prodrug thereof, wherein
- Y is selected from the group consisting of -O-, -NR 5 -, and -S-;
- Z is selected from the group consisting of optionally substituted C1-C6 alkyl, optionally substituted C6-C10 aryl, and optionally substituted C1-C9 heteroaryl, wherein C3-C10 aryl and C1-C9 heteroaryl are optionally substituted with one or more substituents independently selected from the group consisting of -OR' 5 , halo, and C1-C4 alkyl, wherein R 15 is H, C1-C3 alkyl, or C4 alkyl, and/or wherein an alkylene group is attached to two adjacent atoms of the C6-C10 aryl or C1- C9 heteroaryl group to form a 5- or 6-membered partially saturated or saturated ring, optionally wherein the alkylene group is substituted with one or more halo atoms;
- A, B, C and D are each independently selected from the group consisting of H, optionally substituted C1-C6 alkyl, halo, NO 2 , optionally substituted C3-C10 aryl, optionally substituted C1-C9 heteroaryl, -OR 6 , NR 7 R 8 , -SR 9 , -C(O)OR i0 , - C(O)NR r R 12 , -C(O)SR 13 , C(O 2 )SR 14 ;
- R 1 , R 4 , R 5 , R 6 , R 9 , R 10 , R 13 , and R 14 are each independently selected from the group consisting of H and C1-C6 alkyl group, wherein the C1-C6 alkyl is optionally substituted with one or more halo atoms;
- R 2 , R 3 , R 7 , R 8 , R 11 , and R 12 are each independently selected from the group consisting of H and C1-C6 alkyl, wherein the C1-C6 alkyl is optionally substituted with one or more halo atoms, or R 2 and R 3 , R 7 and R 8 , and/or R 11 and R 1Z can be taken together with the nitrogen atom to which they are attached to from a 5- or 6- membered heterocycle, optionally substituted with one or more halo atoms.
- the small molecule sortilin inhibitor may be any of 2-[(6-methylpyridin-2- yi)carbamoyl]benzoic acid, 2-methyl-6-[(6-methylpyridin-2-yl)carbamoyl]benzoic acid, 5- bromo-2-[(6-methylpyridin-2-yl)carbamoyl]benzoic acid, 5-chloro-2-[(6-methylpyridin-2- yl)carbamoyl]benzoic acid, 5-methyl-2-[(6-methylpyridin-2-yl)carbamoyl]benzoic acid, 2-[(6- methylpyridin-2-yl)carbanoyl]-5-(propan-2-yl)benzoic acid 2-[(6-methylpyridin-2- yl)carbamoyl]-5-(trifluoromethyl)benzoic acid, 2-[(6-methylpyridin-2-yl)carbamoyl][(
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2023/101595A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 12 of WO2023/101595A1 , i.e., any of:
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in W02023/031440A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 12 of W02023/031440A1 .
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2023/161505A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 9 of WO2023/161505A1 , i.e., any of (2S)-2- [(benzylcarbamoyl)aminc]-5,5-dimethylhexanoic acid, (2S)-2-
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2024/047227A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 9 or claim 12 of WO2024/047227A1.
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in EP4089102A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 10 of EP4089102A1 .
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2022/223805A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 10 of WO2022/223805A1 .
- the small molecule sortilin inhibitor may also be any of the compounds disclosed in WO2022/157271 A1 , which is hereby incorporated in its entirety by reference.
- the small molecule sortilin inhibitor may be any of the specific compounds recited in claim 9 of WO2022/157271A1.
- sortilin inhibitor may also have a formula of, e.g.: wherein R 1 may be selected from any of:
- the small molecule inhibitor may also have a formula of: wherein R 2 may be selected from any of:
- the small molecule inhibitor of sortilin may have a formula of: wherein R 1 may be selected from any of:
- the small molecule inhibitor of sortilin may also have a formula of: wherein R 1 is and R 2 is phenethyl; or wherein R 1 is and R 2 is benzyl; or wherein R 1 is and R 2 is phenethyl; or wherein and R 2 is benzyl.
- the small molecule inhibitor may also be a sortilin inhibitor as disclosed in Anand et al., 2023, which is herewith incorporated by reference as well.
- the small molecule inhibitor may also be obtainable by a method for identifying a sortilin ligand as disclosed in WO2009132656A2, which is herewith incorporated by reference.
- the sortilin inhibitor may be any of the herein described small molecules.
- the sortilin inhibitor may comprise not only a single type of small molecule but a combination of several different small molecule drugs targeted against sortilin, e.g., it may comprise a combination of any of the herein described small molecule inhibitors such as, e.g., a combination of AF38469 and M PEP, of AF38469 and AF40431 or of AF38469, AF40431 and MPEP.
- the sortilin inhibitor may be an anti-sortilin antibody.
- An antibody also known as immunoglobulin is a natural or synthetic, typically Y-shaped protein that can selectively bind a target antigen.
- the term “antibody” may include both polyclonal and monoclonal antibodies, however, preferably, the anti-sortilin inhibitor is a monoclonal antibody.
- an antibody in the context of the invention may also be a fragment of an antibody, e.g., a Fab fragment or a Fab 2 fragment, a human or humanized antibody, a bivalent antibody or a single chain antibody, as long as it selectively binds the target antigen.
- the antibody may be chosen or designed using standard laboratory techniques to specifically bind to an epitope of sortilin in a fashion that blocks binding of a sortilin ligand to sortilin. Accordingly, in cases where the sortilin inhibitor is an anti-sortilin inhibitor, it preferably functions as an antagonistic antibody.
- AL001 anti-sortilin antibodies
- the first of these antibodies, AL001 is also known under the name Latozinemab. It is disclosed, amongst other anti-sortilin antibodies, in WQ2020014617A1 , which is hereby incorporated by reference.
- AL001 has been successfully tested for the treatment of frontotemporal dementia and/or ALS in Phase 2 and Phase 3 clinical trials, including NCT04374136, NCT03987295, NCT03636204 and NCT05053035.
- AL001 comprises a heavy chain having an amino acid sequence of SEQ ID NO: 1 and a light chain having an amino acid sequence of SEQ ID NO: 2.
- the sortilin inhibitor of the present invention is an anti-sortilin antibody having a heavy chain with an amino acid sequence of SEQ ID NO: 1 and a light chain with an amino acid sequence of SEQ ID NO: 2.
- the second anti-sortilin antibody that tested in a Phase 1 clinical trial (NCT04111666) is known under the name AL101 and is disclosed in WO2022120352A1 , which is herewith incorporated by reference.
- AL101 comprises a light chain variable domain and a heavy chain variable domain, wherein the heavy chain variable domain comprises an HVR-H1 comprising the amino acid sequence of SEQ ID NO:3, an HVR-H2 comprising the amino acid sequence of SEQ ID NO: 4, and an HVR-H3 comprising the amino acid sequence of SEQ ID NO:5, and the light chain variable domain comprises an HVR-L1 comprising the amino acid sequence of SEQ ID NO:6, an HVR-L2 comprising the amino acid sequence of SEQ ID NO: 7, and an HVR-L3 comprising the amino acid sequence of SEQ ID NO:8.
- the anti-sortilin antibody may thus also comprise a light chain comprising the amino acid sequence of SEQ ID NO: 15, and a heavy chain comprising the amino acid sequence of SEQ ID NO: 16 or 17. It can also be used in the context of the invention.
- the anti-sortilin inhibitor may also be any of the other anti-sortilin antibodies besides AL001 that are disclosed in W02020014617A1.
- the anti-sortilin antibody may also be any of the anti-sortilin antibodies disclosed in WO2016/164637A1, which is hereby incorporated in its entirety by reference.
- the sortilin inhibitor may also be an anti-sortilin antibody as disclosed, e.g., in Miyakawa et al., 2020, which is hereby incorporated by reference as well.
- the sortilin inhibitor may comprise a combination of two or more different anti-sortilin antibodies, i.e., a combination of anti-sortilin antibodies that bind to different epitopes of the sortilin protein.
- the sortilin inhibitor may comprise a mixture of the anti-sortilin antibodies AL001 and AL101.
- the sortilin inhibitor may be an inhibitory nucleic acid inhibiting the expression of sortilin. wherein, typically, the inhibitory nucleic acid is an inhibitory RNA.
- RNA interference RNA interference
- RISC RNA-induced silencing complex
- the inhibitory RNA can be, e.g., a short hairpin RNA (shRNA), a microRNA (miRNA) or a small interfering RNA (siFRNA).
- miRNAs are single-stranded, non-coding RNAs that are generated from longer endogenous precursor transcripts forming hairpin structures and are processed through endonucleolytic cleavage into short RNA duplexes of 20-23 base pairs (bp). Typically, only the antisense strand of these duplexes is incorporated into RISC to guide the complex to a complementary target RNA.
- siRNAs Naturally occurring small interfering RNAs (siRNAs) rely on a similar biogenesis pathway as miRNAs but originate from doublestranded RNA molecules rather than from hairpin structures. For instance, in many cases, siRNAs are naturally produced from double-stranded viral RNA. Mature siRNAs typically have a length of 20 to 25 nt and, similar to miRNAs, rely on RNAi to prevent target RNAs from being translated into protein. However, whereas naturally occurring miRNAs typically silence genes by repression of translation and have broader specificity of action, naturally occurring siRNAs typically exhibit a higher specificity as they oftentimes bind with up to 100% sequence complementarity to their target sequences.
- siRNAs thus can induce cleavage of their target mRNA via RISC before translation.
- RISC short hairpin RNAs
- shRNAs typically have a length of 50-70 nt and form a stem-loop structure consisting of a 19 to 29 bp region of double-strand RNA (the stem) bridged by a region of predominantly single-stranded RNA (the loop) and a dinucleotide 3’ overhang.
- the stem double-strand RNA
- the loop a region of predominantly single-stranded RNA
- the dinucleotide 3’ overhang Once they are introduced into a cell, they are processed similar to miRNAs into short RNA duplexes that resemble siRNAs. One strand of this duplex is then incorporated into RISC to facilitate knock-down of a target gene.
- ShRNAs can be encoded by an expression vector, such es a viral vector, which allows expression and processing of the shRNA construct inside a target cell.
- ShRNAs are normally transcribed by RNA polymerase III.
- shRNAs are preferably provided as a "shRNAmiRs”, i.e., the shRNA is embedded into a framework or backbone of a miRNA. Such an shRNAmiR may also be referred to as an artificial miRNA or miRNA mimic.
- shRNAmiRs can be generated from Polymerase II promoters, enabling constitutive, tissue specific or inducible expression (Adams et al., 2017).
- siRNA-mediated knockdown of sortilin has been repeatedly achieved in the literature, e.g. in Lefrancois et al.. 2005; Roselli et al, 2015, Uchiyama et al., 2017 and Gao et al., 2020.
- the sortilin inhibitor is an siRNA
- the siRNA may have a nucleic acid sequence as disclosed in any of these publication, i.e.
- the siRNA targeted against sortilin may comprise or consist of a nucleic acid sequence of any of, e.g., SEQ ID NOs: 9-14 (9: AGGTGGTGTTAACAGCAGAG, 10: AATGTTCCAATGCCCCACTC, 11 : CUCUGCUGUUAACACCACC[dT][dT], 12:
- the sortilin inhibitor is an inhibitory RNA
- the RNA may comprise at least one modification to increase its stability.
- the skilled person is aware of various such stabilizing modifications that may be introduced into, e.g., an siRNA. Suitable modifications are summarized e.g., in Selvam et al., 2017.
- the skilled person may choose to design an inhibitory nucleic acid that is not necessarily an RNA, e.g., it may be an antisense oligonucleotide targeted against the mRNA of sortilin to prevent its expression.
- Antisense oligonucleotides are short synthetic nucleic acids made either from DNA or RNA that sequence-specifically bind to target mRNAs via complementary base pairing to block their translation into protein. As in the case of inhibitory RNAs for use in RNAi-mediated gene knockdown, a person skilled in the art is aware of standard laboratory techniques well known to the field to choose or design appropriate antisense oligonucleotides that specifically target the mRNA of sortilin.
- the inhibitory nucleic acid may also be an aptamer against sortilin, i.e., an artificial oligonucleotide made of DNA, RNA or XNA that, due to its specific 3-dimensional structure, can bind with high affinity to the sortilin protein to cause its inhibition.
- XNA short for xenobiotic nucleic acid refers to a synthetic nucleic acid analogue. The skilled person will be aware of types of XNAs suitable for aptamer production.
- the XNA-aptamer may consist of 5-anhydrohexitol nucleic acid (HNA).
- the aptamer may be an L- ribonucleic acid aptamer (also known by its tradename Spiegelmer).
- Spiegelmers are artificial RNA oligonucleotides built of L-ribose rather than D-ribose, thus rendering them highly resistant to degradation by nucleases. Like conventional aptamers, Spiegelmers directly bind to their target proteins to mediate their functional inhibition.
- the sortilin inhibitor is a single type of inhibitory nucleic acid, e.g., it may be an siRNA having or comprising any of the sequences disclosed herein.
- the sortilin inhibitor may be a composition comprising a mixture of different inhibitory nucleic acids.
- the sortilin inhibitor for the herein described use may comprise an inhibitory RNA, e.g., one or more the siRNA molecules described herein as well as an antisense oligonucleotide directed to sortilin mRNA.
- the sortilin inhibitor may also comprise a mixture of different siRNA constructs directed to sortilin mRNA, e.g., the sortilin inhibitor may comprise a mixture of siRNAs of any of the herein described SEQ ID NO.: 9-14.
- the sortilin inhibitor may be a peptide-based inhibitor, i.e., it may be a therapeutic peptide that is composed of a series of amino acids. It may have a molecular weight that is bigger than that of a small molecule as defined herein. Accordingly, a peptide- based inhibitor within the meaning of the present invention typically has a molecular weight of more than 800 Dalton, e.g., of 800 to 10,000 Dalton or 2000 to 5000 Dalton.
- the term “peptide-based inhibitor” may also refer to a soluble fragment of sortilin, or a sortilin-related molecule, which can bind competitively to biologically available sortilin ligands such as pro- neurotrophins. As a result, the sortilin inhibitor results in an inhibition of receptor activation, which would otherwise occur as a result of ligand binding.
- the sortilin inhibitor may also be proteolysis-targeting chimera (PROTAC).
- PROTACs consist of two different protein-binding molecules that are covalently connected to each other via short linker region. One of the two protein-binding molecules is capable of binding to a target protein meant for degradation, i.e., in the present case to sortilin. The second molecule binds to an E3 ubiquitin ligase. Accordingly, PROTACs are able to bring the target protein and the E3 ligase into close proximity so that the target protein can be ubiquitinated and thus marked for proteasomal degradation. The skilled person will be able to develop effective PROTACs against sortilin based on the known prior art.
- PROTACs need only bind to a target protein with high selectivity, without, however, having to exert an inhibitory effect on the enzymatic activity of said target protein.
- an antibody to sortilin or a derivative thereof, e.g., a scFv may be used.
- the sortilin inhibitor may also be a combination of any of the different types of inhibitors described herein.
- the sortilin inhibitor may comprise a mixture of an anti-sortilin antibody such as AL001 or AL101 and/or a small molecule inhibitor such as AF38469.
- the condition associated with the release of an endocrine and/or a paracrine factor by a functional neuroendocrine tumor is carcinoid syndrome
- the endocrine and/or a paracrine factor is serotonin
- the sortilin inhibitor is 2- [[(6-methyl-2-pyridinyl)amino]carbonyl]-5-(trifluoromethyl)-benzoic acid or an anti-sortilin antibody having a heavy chain with an amino acid sequence of SEQ ID NO: 1 and a light chain with an amino acid sequence of SEQ ID NO: 2.
- it is 2-[[(6-methyl-2- pyridinyl)amino]carbonyl]-5-(trifluoromethyl)-benzoic acid.
- the sortilin inhibitor may be provided in a suitable pharmaceutical composition, wherein said pharmaceutical composition may comprise additional ingredients such as, e.g., a pharmaceutically acceptable excipient.
- pharmaceutically acceptable refers those compounds, materials, compositions, and/or dosage forms that are, within the scope of sound medical judgement, suitable for use in contact with the tissues of the subject without causing excessive toxicity, irritation, allergic response or other problems or complications commensurate with a reasonable benefit/risk ratio.
- the pharmaceutical composition further comprises a suitable carrier, i.e., a compound, composition, structure or substance that, when in combined with the sortilin inhibitor described herein, aids or facilitates preparation, storage, administration, delivery, effectiveness, selectivity, or any other feature of the inhibitor for its intended use or purpose.
- a suitable carrier i.e., a compound, composition, structure or substance that, when in combined with the sortilin inhibitor described herein, aids or facilitates preparation, storage, administration, delivery, effectiveness, selectivity, or any other feature of the inhibitor for its intended use or purpose.
- a suitable carrier i.e., a compound, composition, structure or substance that, when in combined with the sortilin inhibitor described herein, aids or facilitates preparation, storage, administration, delivery, effectiveness, selectivity, or any other feature of the inhibitor for its intended use or purpose.
- It may e.g. comprise water, a saline, e.g., physiological saline, or a buffer.
- said carrier is a vector comprising or encoding the sortilin inhibitor.
- the type of vector depends primarily on the nature of the inhibitor used to inhibit sortilin.
- the sortilin inhibitor is a small molecule inhibitor, an anti-sortilin antibody, an in-vitro generated inhibitory nucleic acid, a PROTAC or a peptide-based inhibitor
- the vector used for transporting the sortilin inhibitor to its site of action may be a lipid vesicle or comprise a lipid membrane that encapsulates the inhibitor.
- the vector may be a liposomal or an exosomal carrier comprising the sortilin inhibitor as a cargo and, optionally, expressing a targeting moiety on its surface.
- the vector may also be a lipid nanoparticle.
- the vector carrying the sortilin inhibitor may be a bacterial minicell. These types of carriers have the ability to entrap both hydrophilic therapeutic agents within their central aqueous core or lipophilic drugs within their bilayer compartment.
- the targeting moiety may be, e.g., a cell-specific antibody or ligand such as, e.g., a small peptide, polysaccharide or nucleic acid that can be bound by a receptor expressed on the surface of the cell of interest.
- the vector may be a polymeric micro- or nanoparticle.
- the micro- or nanoparticle may be formed of any physiologically acceptable polymeric material known in the art, e.g., it may comprise a biopolymer such as polysaccharide selected from the group comprising cellulose, alginate or chitosan.
- the vector may also be a nanocarrier comprising, e.g., a hyperbranched dendritic polyglycerol polymer.
- a nanocarrier is an amphiphilic unimolecular nanocarrier of dendritic structure, and it comprises a dendritic core and at least one shell.
- the sortilin inhibitor is an inhibitory nucleic acid that is not in vitro-transcribed, it is inserted into an express on vector encoding the inhibitory nucleic acid.
- expression vectors include but are not limited to minicircles, plasmids, cosmids, phages, viruses or artificial chromosomes.
- the vector preferably is a viral vector, e.g., a lentiviral vector, an adenoviral vector or an adeno-associated viral vector.
- Expression vectors typically contain a cargo sequence comprising an expression cassette, i.e. the necessary elements that permit transcription of a nucleic acid into inhibitory nucleic acid, such as a suitable promoter.
- a stretch of cargo DNA, i.e. , a transgene, encoding an nhibitory RNA may be operably linked to a suitable promoter and inserted into the expression vector of choice.
- the promoter used to express the inhibitory RNA may be any constitutively active promoter known in the art, e.g., a Rous sarcoma virus (RSV) promoter, a human cytomegalovirus (CMV) promoter, an HIV promoter or a eukaryotic promoter such as e.g...
- the promoter may be an inducible promoter that only drives expression of the inhibitory RNA in the presence or absence of a certain stimulus.
- the inducible promoter may be a Tet-regulated promoter, i.e., it may comprise a tetracycline response element (TRE) that can be bound by a tetracycline transactivator (tTA) protein in the presence of tetracycline or an analogue thereof, e.g. doxycycline.
- TRE tetracycline response element
- tTA tetracycline transactivator
- the promoter may also be a cell-specific promoter which only drives expression in a particular type of cell, in the present case, in the NET of interest.
- the assembled viral vector is preferably pseudotyped, e.g., with a viral envelope glycoprotein or a capsid capable of binding to a receptor expressed on the surface of the cell of interest.
- pseudotyping refers to the generation of viral vectors that carry foreign viral envelope glycoproteins on their surface or are encapsulated in a capsid of a different virus or viral serotype to specifically target only particular cell types of interest.
- the viral vector particles are usually assembled inside suitable packaging cells known in the art according to standard laboratory techniques before they are administered to the subject.
- Inhibitory nucleic acids may also be delivered to the cells of interest using a vector that is a non-pathogenic, genetically modified bacterium or yeast comprising a prokaryotic or eukaryotic vector.
- the vector comprises a DNA molecule encoding the inhibitory nucleic operably linked to a promoter that controls expression of said inhibitory nucleic acid, as described above.
- the ncn-pathogenic bacterium is preferably invasive, i.e., it is capable of entering a host cell and may be derived, e.g., from a non-pathogenic strain of Escherichia coll, Listeria, Yersinia, Rickettsia, Shigella, Salmonella, Legionella, Chlamydia, Brucella, Neisseria, Burkolderia, Bordetella, Borrelia, Coxiella, Mycobacterium, Helicobacter, Staphylococcus, Streptococcus, Vibrio, Lactobacillus, Porphyromonas, Treponema, or Bifidobacteriae.
- the prokaryotic or eukaryotic vector is specifically designed for targeted delivery to a cell of interest, e.g., by expressing a ligand on its surface that can be recognized by a receptor on the target cell.
- the inhibitory nucleic is not expressed on site from an expression vector at the cell of interest, but instead is expressed in vitro before being packaged into a suitable vector vehicle, e.g., as described above, and directly delivered to the target cell. This way, the time of nucleic acid-mediated target gene inhibition can be effectively limited.
- the sortilin inhibitor according to the invention may be administered directly to the subject in need thereof, e.g., orally, intravenously, intramuscularly, intraperitoneally, subcutaneously, transdermally or transmucosally. Administration may be by a single administration, e.g., an injection or infusion. A slow-reiease formulation may also be chosen if administration over a longer time period is desired. Oral administration may be in a form suitable for targeted release in a specific area of the gut, e.g., depending on the location of the NET.
- a therapeutically effective amount of the sortilin inhibitor is to be administered to the subject.
- a therapeutically effective’ amount refers to the amount of the inhibitor that is of sufficient quantity to ameliorate one or more causes or symptoms of a condition as described herein. Such amelioration only requires a reduction or alteration, not necessarily elimination.
- a person skilled in the art will be able to choose a suitable vector depending on the type of inhibitor used, the route of administration, the respective target cell to which the inhibitor is to be delivered and the condition to be treated. In addition, based on the known state of the art, the skilled person will be able to further modify the vector for optimized targeted delivery and release of the inhibitor.
- Fig. 1 Sortilin is a marker of functional neuroendocrine tumors (NETs).
- A Representative images of a pancreatic NET stained for sortilin (top panels) or by using secondary antibody only (control, bottom panels)
- B Representative images of a small intestinal NET stained to' sortilin (top panels) or by using secondary antibody only (control, bottom panels)
- C Representative images of a small intestinal NET stained to' sortilin (top panels) or by using secondary antibody only (control, bottom panels)
- C)-(F) Staining intensity was estimated as described in Kim et al.
- Fig. 2 Sortilin is expressed in neuroendocrine tumor cell cultures. Western blot for assessing sortilin expression in non-neuroendocrine (Hela) cell culture as well as two neuroendocrine tumor cell cultures (QGP1 and BON).
- Fig. 3 Concentration of amines in BON cell lysates and supernatants after treatment with sortilin inhibitor
- (A)-(D) BON neuroendocrine tumor cells were incubated for 23h in serotonin free medium with (Sort Inh) or without (Control) the sortilin inhibitor AF38469 (10pM). Afterwards, the serotonin (5HT, A), Tryptophan (TRP, B) and Tyrosin (TYR, C) content as well as the content of the serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) in the cell culture supernatant was assessed using high performance liquid chromatography (HPLC). The cells were treated with fresh sortilin inhibitor for two more hours. Subsequently, the 5HT, (E, F), TRP.
- HPLC high performance liquid chromatography
- Fig. 4 Proliferation of BON cells after sortilin inhibition. To exclude the possibility of toxic effects of sortilin inhibition on BON cells, a cell proliferation assay was performed using the Alamar Blue cell viability assay 5 days after sortilin inhibition.
- Fig. 5 Sortilin is expressed in healthy human tissues.
- A Western blot analysis demonstrates expression of sortilin protein in healthy human colon and ileum tissues as well as in a functional neuroendocrine tumor.
- B Representative image of healthy human pancreas tissue immunostained for sortilin.
- C Representative image of healthy human colon tissue immunostained for sortilin. Immunostaining revealed that the endocrine part of the pancreas (the pancreatic islet) was highly positive for sortilin. Staining the midgut for sortilin revealed scattered sortilin positive cells resembling neuroendocrine cells.
- Fig. 6 Sortilin is expressed in neuroendocrine cells.
- Fig. 7 Effect of sortilin inhibition on serotonin content of neuroendocrine organoids.
- A Experimental scheme for testing the effects of sortilin inhibition on serotonin levels in neuroendocrine organoids.
- Murine intestinal organoids are generated and enriched with neuroendocrine cells as previously described in (Kawasaki et al., 2018; Basak et al., 2017). Enrichment with neuroendocrine cell is demonstrated by immunostaining for synaptophysin.
- Organoids are subsequently incubated four days in the absence (control) or the presence of the sortilin inhibitor AF38469 (10pM). The lysates of these organoids are next harvested, and serotonin levels are measured by HPLC.
- (B) Organoids treated with the sortilin inhibitor (Sort Inh) showed a lower concentration of serotonin (5HT) per cell. This result underlines the role of sortilin in hormone production in neuroendocrine cells and underpins sortilin as a promising therapeutic target.
- Fig. 8 Impact of sortilin deficiency on splenic amine concentration in sortilin knockout mice.
- a sortilin knockout (KO) mouse model was generated previously (Jansen et al., 2007) by deleting 41 codons in exon 14 of the Sortl gene.
- Sortilin KO mice exhibited reduced serotonin (5HT, A), Tryptophan (Trp, B) and Tyrosine (Tyr, C) levels in their spleens compared to wild type (WT) mice.
- Sortilin also known as neurotensin receptor 3 is a member of the VPS10P domain sorting receptor family, a group of transmembrane receptors which are involved in sorting and trafficking of a broad range of ligands (Malik et al., 2020). It has been already shown that sortilin is expressed in many healthy as well as in many cancer cells like breast cancer or lung cancer (Roselli et al., 2015; Al Akhrass et al., 2017; Gao et al., 2018). Expression was also found in NETs and was attributed to cell migration and adhesion (Kim et al., 2018). However, only a fraction of the stained tissue samples was positive for sortilin expression (Kim et al., 2018).
- NET samples for sortilin used antibody: R&D systems: AF3154
- Fig. 1A, B a total of 21 intestinal or pancreatic primary tumors and 31 hepatic metastases of intestinal, pancreatic or lung (1 sample) NETs were analyzed. All immunostained primary tumors also had hepatic metastases. Staining intensity was estimated by an immunoreactivity score, which was defined by the product of the value for immunostaining intensity (0, no staining; 1 , weak staining; 2, moderate staining; 3, strong staining) and for the percentage of positive immunoreactive cells (0, no positive; 1 , 0%-10% positive; 2, 11 %-50% positive; 3, 51 %-100% positive).
- sortilin is a marker of functional NETs. To our knowledge this finding represents the first marker which distinguishes functional from nonfunctional tumors.
- sortilin has a functional role in hormone production or secretion of neuroendocrine tumors.
- the serotonin producing and sortilin expressing neuroendocrine BON cell culture Karl et al., 2018: Samuel Tran et al., 2004.
- Fig. 5A Immunostaining revealed that the endocrine part of the pancreas (the pancreatic islet) was highly positive for sortilin (Fig. 5B). Staining the midgut for sortilin revealed scattered sortilin positive cells resembling neuroendocrine cells (Fig. 5C). Double immunostaining for synaptophysin and sortilin could validate this assumption (Fig. 6A and B). This is in line with previous published studies, which could observe sortilin in neuroendocrine cells (Boggild et al., 2016).
- sortilin KO mice were obtained by homologous recombination, thereby deleting 41 codons in exon 14 of the Sortl gene. Mice homozygous for this deletion are viable and do not show apparent clinical alterations. Although not significant, we found that sortilin KO mice exhibited reduced 5HT levels in the spleen, which is indicative of a lower 5HT concentration in the blood (Fig. 8). As these mice lack completely sortilin starting already from the embryonic state on without an apparent phenotype, it is expected that compensatory mechanisms took place in order to attenuate the effect of the sortilin knock-out.
- sortilin represents a completely new target for the treatment of the functionality (e.g. carcinoid syndrome) of neuroendocrine tumors.
- sortilin a novel lysosomal sorting receptor
- Halperin DM Shen C, Dasari A, Xu Y, Chu Y, Zhou S, et al. Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study. The Lancet Oncology. 2017; 18(4):525-34.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Medicinal Chemistry (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Organic Chemistry (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
La présente invention appartient au domaine des thérapies anticancéreuses, et concerne plus particulièrement des thérapies visant à améliorer la santé de patients souffrant de tumeurs neuroendocrines (NET) fonctionnelles ou de néoplasmes neuroendocriniens (NEN) fonctionnels. En particulier, l'invention divulgue l'utilisation d'inhibiteurs de sortiline pour traiter une affection associée à la libération d'un facteur endocrinien et/ou paracrine tel que la sérotonine par un néoplasme neuroendocrinien fonctionnel chez un sujet, par exemple le syndrome carcinoïde, une maladie cardiaque carcinoïde et/ou une crise carcinoïde. L'inhibiteur de sortiline peut être fourni dans une composition pharmaceutique et peut être un inhibiteur à petite molécule ayant une masse moléculaire inférieure ou égale à 800 daltons, un anticorps anti-sortiline, un acide nucléique inhibiteur, une chimère ciblant la protéolyse, un inhibiteur à base de peptide ou une combinaison de ceux-ci.
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| EP23175836.8 | 2023-05-26 | ||
| EP23175836 | 2023-05-26 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024245719A1 true WO2024245719A1 (fr) | 2024-12-05 |
Family
ID=86605642
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/EP2024/062984 Pending WO2024245719A1 (fr) | 2023-05-26 | 2024-05-10 | Inhibiteurs de sortiline pour le traitement de patients atteints de tumeurs neuroendocrines fonctionnelles |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2024245719A1 (fr) |
Citations (13)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2009132656A2 (fr) | 2008-04-27 | 2009-11-05 | H. Lundbeck A/S | Mise au point de ligands spécifiques de la sortiline |
| WO2016164637A1 (fr) | 2015-04-07 | 2016-10-13 | Alector Llc | Anticorps anti-sortiline et leurs méthodes d'utilisation |
| WO2019040497A1 (fr) * | 2017-08-22 | 2019-02-28 | Scripps Health | Procédé de traitement d'une tumeur neuroendocrine |
| WO2020014617A1 (fr) | 2018-07-13 | 2020-01-16 | Alector Llc | Anticorps anti-sortiline et leurs méthodes d'utilisation |
| WO2021186054A1 (fr) | 2020-03-19 | 2021-09-23 | INSUSENSE ApS | Antagonistes de la sortiline pour destinés à être utilisés dans le traitement de la rétinopathie diabétique |
| WO2022120352A1 (fr) | 2020-12-02 | 2022-06-09 | Alector Llc | Méthodes d'utilisation d'anticorps anti-sortiline |
| WO2022157271A1 (fr) | 2021-01-20 | 2022-07-28 | INSUSENSE ApS | Dérivés de pyridine en tant que modulateurs de l'activité de la sortiline |
| WO2022223805A1 (fr) | 2021-04-23 | 2022-10-27 | INSUSENSE ApS | Modulateurs de l'activité de la sortiline |
| EP4089102A1 (fr) | 2021-05-14 | 2022-11-16 | Insusense ApS | Modulateurs de la activité de sortiline |
| WO2023031440A1 (fr) | 2021-09-03 | 2023-03-09 | INSUSENSE ApS | Dérivés d'acide 2-amino-5,5-diméthylhexanoïque comme modulateurs de sortiline pour utilisation dans le traitement des maladies du système nerveux central |
| WO2023101595A1 (fr) | 2021-12-02 | 2023-06-08 | Sortina Pharma Ab | Inhibiteurs de la sortiline |
| WO2023161505A1 (fr) | 2022-02-28 | 2023-08-31 | INSUSENSE ApS | Carbamates et/ou urées à base d'acides aminés pour le traitement de maladies dépendantes de la sortiline |
| WO2024047227A1 (fr) | 2022-09-02 | 2024-03-07 | INSUSENSE ApS | Modulateurs de sortiline |
-
2024
- 2024-05-10 WO PCT/EP2024/062984 patent/WO2024245719A1/fr active Pending
Patent Citations (13)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| WO2009132656A2 (fr) | 2008-04-27 | 2009-11-05 | H. Lundbeck A/S | Mise au point de ligands spécifiques de la sortiline |
| WO2016164637A1 (fr) | 2015-04-07 | 2016-10-13 | Alector Llc | Anticorps anti-sortiline et leurs méthodes d'utilisation |
| WO2019040497A1 (fr) * | 2017-08-22 | 2019-02-28 | Scripps Health | Procédé de traitement d'une tumeur neuroendocrine |
| WO2020014617A1 (fr) | 2018-07-13 | 2020-01-16 | Alector Llc | Anticorps anti-sortiline et leurs méthodes d'utilisation |
| WO2021186054A1 (fr) | 2020-03-19 | 2021-09-23 | INSUSENSE ApS | Antagonistes de la sortiline pour destinés à être utilisés dans le traitement de la rétinopathie diabétique |
| WO2022120352A1 (fr) | 2020-12-02 | 2022-06-09 | Alector Llc | Méthodes d'utilisation d'anticorps anti-sortiline |
| WO2022157271A1 (fr) | 2021-01-20 | 2022-07-28 | INSUSENSE ApS | Dérivés de pyridine en tant que modulateurs de l'activité de la sortiline |
| WO2022223805A1 (fr) | 2021-04-23 | 2022-10-27 | INSUSENSE ApS | Modulateurs de l'activité de la sortiline |
| EP4089102A1 (fr) | 2021-05-14 | 2022-11-16 | Insusense ApS | Modulateurs de la activité de sortiline |
| WO2023031440A1 (fr) | 2021-09-03 | 2023-03-09 | INSUSENSE ApS | Dérivés d'acide 2-amino-5,5-diméthylhexanoïque comme modulateurs de sortiline pour utilisation dans le traitement des maladies du système nerveux central |
| WO2023101595A1 (fr) | 2021-12-02 | 2023-06-08 | Sortina Pharma Ab | Inhibiteurs de la sortiline |
| WO2023161505A1 (fr) | 2022-02-28 | 2023-08-31 | INSUSENSE ApS | Carbamates et/ou urées à base d'acides aminés pour le traitement de maladies dépendantes de la sortiline |
| WO2024047227A1 (fr) | 2022-09-02 | 2024-03-07 | INSUSENSE ApS | Modulateurs de sortiline |
Non-Patent Citations (34)
| Title |
|---|
| AI-AKHRASS HNAVES TVINCENT FMAGNAUDEIX ADURAND KBERTIN F ET AL.: "Sortilin limits EGFR signaling by promoting its internalization in lung cancer", NATURE COMMUNICATIONS, vol. 8, no. 1, 2017, pages 1182, XP055671860, DOI: 10.1038/s41467-017-01172-5 |
| ANAND ET AL.: "Discovery of novel small molecule inhibitors targeting progranulin-sortilin", A VIRTUAL HIGH THROUGHPUT SCREENING APPROACH, 2023 |
| BASAK OBEUMER JWIELBRANDS KSENO HVAN OUDENAARDEN ACLEVERS H: "Induced Quiescence of Lgr5+ Stem Cells in Intestinal Organoids Enables Differentiation of Hormone-Producing Enteroendocrine Cells", CELL STEM CELL, vol. 20, no. 2, 2017, pages 177 - 90 |
| BOGGILD SMOLGAARD SGLERUP SNYENGAARD JR: "Spatiotemporal patterns of sortilin and SorCS2 localization during organ development", BMC CELL BIOL., vol. 17, 2016, pages 8 |
| BOLDUAN FELIX ET AL: "Elevated sortilin expression discriminates functional from non-functional neuroendocrine tumors and enables therapeutic targeting", FRONTIERS IN ENDOCRINOLOGY, vol. 15, 17 April 2024 (2024-04-17), CH, XP093181058, ISSN: 1664-2392, DOI: 10.3389/fendo.2024.1331231 * |
| CLEMENT DRAMAGE JSRIRAJASKANTHAN R: "Update on Pathophysiology, Treatment, and Complications of Carcinoid Syndrome", JOURNAL OF ONCOLOGY, vol. 2020, 2020, pages 8341426 |
| CONTRACTOR TCLAUSEN RHARRIS GRROSENFELD JACARPIZO DRTANG L ET AL.: "IGF2 drives formation of ileal neuroendocrine tumors in patients and mice", ENDOCRINE-RELATED CANCER, vol. 27, no. 3, 2020, pages 175 - 86 |
| DASARI ASHEN CHALPERIN DZHAO BZHOU SXU Y ET AL.: "Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine Tumors in the United States", JAMA ONCOLOGY, vol. 3, no. 10, 2017, pages 1335 - 42 |
| FELIX F. ADAMS, DIRK HECKL, THOMAS HOFFMANN, STEVEN R. TALBOT, ARNOLD KLOOS, FELICITAS THOL, MICHAEL HEUSER, JOHANNES ZUBER, AXEL : "An optimized lentiviral vector system for conditional RNAi and efficient cloning of microRNA embedded short hairpin RNA libraries", BIOMATERIALS, vol. 139, 2017, pages 102 - 115, XP085076814, ISSN: ISSN 0142-9612, https://doi.org/10.1016/j.biomaterials.2017.05.032, DOI: 10.1016/j.biomaterials.2017.05.032 |
| GAO FGRIFFIN NFAULKNER SLI XKING SJJOBLING PDENHAM JWJIANG CCHONDERMARCK H.: "The Membrane Protein Sortilin Can Be Targeted to Inhibit Pancreatic Cancer Cell Invasion", AM J PATHOL, vol. 190, no. 9, 8 June 2020 (2020-06-08), pages 1931 - 1942 |
| GAO FGRIFFIN NFAULKNER SLI XKING SJJOBLING PDENHAM JWJIANG CCHONDERMARCK H.: "The Membrane Protein Sortilin Can Be Targeted to Inhibit Pancreatic Cancer Cell Invasion", AM J PATHOL., vol. 190, no. 9, 8 June 2020 (2020-06-08), pages 1931 - 1942 |
| GAO FGRIFFIN NFAULKNER SROWE CWWILLIAMS LROSELLI S ET AL.: "The neurotrophic tyrosine kinase receptor rrka and its ligand NGF are increased in squamous cell carcinomas of the lung", SCI REP, vol. 8, no. 1, 2018, pages 8135 |
| GUILLAUMIE K ET AL.: "Serotonin Secretion by Human Carcinoid BON Cells", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, vol. 1014, no. 1, 2004, pages 179 - 88, XP071400398, DOI: 10.1196/annals.1294.019 |
| HALPERIN DMSHEN CDASARI AXU YCHU YZHOU S ET AL.: "Frequency of carcinoid syndrome at neuroendocrine tumour diagnosis: a population-based study", LANCET ONCOLOGY., vol. 18, no. 4, 2017, pages 525 - 34, XP029962214, DOI: 10.1016/S1470-2045(17)30110-9 |
| ITO TLEE LJENSEN RT: "Carcinoid-syndrome: recent advances, current status and controversies", CURR OPIN ENDOCRINOL DIABETES OBES, vol. 25, no. 1, February 2018 (2018-02-01), pages 22 - 35 |
| JANSEN PGIEHL KNYENGAARD JRTENG KLIOUBINSKI OSJOEGAARD SS ET AL.: "Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury", NAT NEUROSCI., vol. 10, no. 11, 2007, pages 1449 - 57, XP002478373, DOI: 10.1038/nn2000 |
| JOISH VNSHAH STIERCE JCPATEL DMCKEE CLAPUERTA P ET AL.: "Serotonin levels and 1-year mortality in patients with neuroendocrine tumors: a systematic review and meta-analysis", FUTURE ONCOLOGY, vol. 15, no. 12, 2019, pages 1397 - 406 |
| KAWASAKI KFUJII MSATO T: "Gastroenteropancreatic neuroendocrine neoplasms: genes, therapies and models", DISEASE MODELS & MECHANISMS, vol. 11, no. 2, 2018 |
| KAWASAKI KTOSHIMITSU KMATANO MFUJITA MFUJII MTOGASAKI K ET AL.: "An Organoid Biobank of Neuroendocrine Neoplasms Enables Genotype-Phenotype Mapping", CELL, vol. 183, no. 5, 2020, pages 1420 - 35, XP086368300, DOI: 10.1016/j.cell.2020.10.023 |
| KIM JI TAE ET AL: "Neurotensin Receptor 3/Sortilin Contributes to Tumorigenesis of Neuroendocrine Tumors Through Augmentation of Cell Adhesion and Migration", NEOPLASIA, vol. 20, no. 2, 1 February 2018 (2018-02-01), US, pages 175 - 181, XP093181083, ISSN: 1476-5586, DOI: 10.1016/j.neo.2017.11.012 * |
| KIM JTNAPIER DLWEISS HLLEE EYTOWNSEND CMJR., EVERS BM.: "Neurotensin Receptor 3/Sortilin Contributes to Tumorigenesis of Neuroendocrine Tumors Through Augmentation of Cell Adhesion and Migration", NEOPLASIA, vol. 20, no. 2, 2018, pages 175 - 81 |
| LEE WCALMEIDA SPRUDENCIO MCAULFIELD TRZHANG YJTAY WMBAUER POCHEW JSASAGURI HJANSEN-WEST KR: "Targeted manipulation of the sortilin-progranulin axis rescues progranulin haploinsufficiency", HUM MOL GENET, vol. 23, no. 6, 26 October 2013 (2013-10-26), pages 1467 - 78, XP055193340, DOI: 10.1093/hmg/ddt534 |
| LEE WCALMEIDA SPRUDENCIO MCAULFIELD TRZHANG YJTAY WMBAUER POCHEW JSASAGURI HJANSEN-WEST KR: "Targeted manipulation of the sortilin-progranulin axis rescues progranulin haploinsufficiency", HUM MOL GENET., vol. 23, no. 6, 26 October 2013 (2013-10-26), pages 1467 - 78, XP055193340, DOI: 10.1093/hmg/ddt534 |
| LEFRANCOIS, S.CANUEL, M.ZENG, J ET AL.: "Inactivation of sortilin (a novel lysosomal sorting receptor) by dominant negative competition and RNA interference", BIOL. PROCED. ONLINE, vol. 7, 2005, pages 17 - 25, Retrieved from the Internet <URL:https:l/doi.org/10.1251/bp101> |
| MALIK ARWILLNOW TE: "VPS10P Domain Receptors: Sorting Out Brain Health and Disease", TRENDS NEUROSCI., vol. 43, no. 11, 2020, pages 870 - 85, XP086321701, DOI: 10.1016/j.tins.2020.08.003 |
| MIYAKAWA SSAKUMA HWARUDE DASANUMA SARIMURA NYOSHIHARA TTAVARES DHATA AIDA KHORI Y: "Anti-sortilin1 Antibody U p-Regulates Progranulin via Sortilin1 Down-Regulation", FRONT NEUROSCI., vol. 14, 15 December 2020 (2020-12-15), pages 586107 |
| RINDI GWIEDENMANN B: "Neuroendocrine neoplasia of the gastrointestinal tract revisited: towards precision medicine", NATURE REVIEWS ENDOCRINOLOGY, vol. 16, no. 10, 2020, pages 590 - 607, XP037241493, DOI: 10.1038/s41574-020-0391-3 |
| ROSELLI S., PUNDAVELA J., DEMONT Y., FAULKNER S., KEENE S., ATTIA J., JIANG C. CHEN, ZHANG X. DONG, WALKER R4. M., HONDERMARCK H: "Sortilin is associated with breast cancer aggressiveness and contributes to tumor cell adhesion and invasion", ONCOTARGET., vol. 6, no. 12, 2015, pages 10473 - 10486, XP055542533, DOI: 10.18632/oncotarget.3401 |
| S, CANUEL MZENG JMORALES CR: "Inactivation of sortilin (a novel lysosomal sorting receptor) by dominant negative competition and RNA interference", BIOL PROCED, vol. 7, 25 January 2005 (2005-01-25), pages 17 - 25 |
| SCHRØDER TJCHRISTENSEN SLINDBERG SLANGGÅRD MDAVID LMALTAS PJESKILDSEN JJACOBSEN JTAGMOSE LSIMONSEN KB: "The identification of AF38469: an orally bioavailable inhibitor of the VPS10P family sorting receptor Sortilin", BIOORG MED CHEM LETT., vol. 24, no. 1, 27 November 2013 (2013-11-27), pages 177 - 80, XP055379658, DOI: 10.1016/j.bmcl.2013.11.046 |
| SELVAM CMUTISYA DPRAKASH SRANGANNA KTHILAGAVATHI R: "Therapeutic potential of chemically modified siRNA: Recent trends", CHEM BIOL DRUG DES, vol. 90, no. 5, 16 May 2017 (2017-05-16), pages 665 - 678, XP072379299, DOI: 10.1111/cbdd.12993 |
| STACHEL ET AL.: "Identification of potent inhibitors of the sortilin-progranulin interaction", BIOORG MED CHEM LETT., 2020 |
| UCHIYAMA KTOMITA MYANO MCHIDA JHARA HDAS NRNYKJAER ASAKAGUCHI S: "Prions amplify through degradation of the VPS10P sorting receptor sortilin", PLOS PATHOG, vol. 13, no. 6, 30 June 2017 (2017-06-30), pages 1006470 |
| WIEDENMANN BJOHN MAHNERT-HILGER GRIECKEN E-O: "Molecular and cell biological aspects of neuroendocrine tumors of the gastroenteropancreatic system", JOURNAL OF MOLECULAR MEDICINE., vol. 76, no. 9, 1998, pages 637 - 47 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US8007790B2 (en) | Methods for treating polycystic kidney disease (PKD) or other cyst forming diseases | |
| CN114144423B (zh) | 使用与免疫检查点抑制剂组合递送的siRNA沉默TGF-BETA 1和COX2以治疗癌症 | |
| JP7536049B2 (ja) | 血管新生障害の処置 | |
| WO2021046225A1 (fr) | Procédés et compositions pour le traitement du cancer | |
| JP2025011102A (ja) | 肝疾患を治療するための組成物および方法 | |
| Umbarkar et al. | Repurposing Nintedanib for pathological cardiac remodeling and dysfunction | |
| WO2012064967A2 (fr) | Le récepteur activateur du ligand nf-kb, issu de cellules cancéreuses, entraîne des métastases des os et des tissus mous | |
| US20130263297A1 (en) | Methods of treating cancer | |
| WO2024245719A1 (fr) | Inhibiteurs de sortiline pour le traitement de patients atteints de tumeurs neuroendocrines fonctionnelles | |
| CN112353940B (zh) | 一种用于预防或治疗抑郁症的药物及其应用 | |
| CN112007157B (zh) | Mrg15蛋白或基因作为靶点在代谢疾病治疗和预防中的应用 | |
| WO2021075559A1 (fr) | Inhibiteur de croissance cellulaire ou inducteur de mort cellulaire pour fibroblastes associés au cancer | |
| US20210024937A1 (en) | Methods of Modulating Lymphangiogenesis, E.g., to Treat Corneal Transplant Rejection, in a Subject | |
| US20220193113A1 (en) | Novel therapeutic agent for gastrointestinal cancer and method for screening for the same | |
| JP2025521567A (ja) | 胆汁うっ滞及び/又は胆汁うっ滞に関連する線維症の予防、進行の遅延又は治療のための方法 | |
| WO2024204685A1 (fr) | Composition pharmaceutique pour le traitement et/ou la prévention du cancer | |
| CN116173040A (zh) | Xmd17109作为arih1激动剂的用途 | |
| JP2014105177A (ja) | 糖尿病治療用医薬組成物及びその有効成分のスクリーニング方法 | |
| HK40023089A (en) | Method for identifying nucleic acids regulating metastasation | |
| HK1245330B (en) | Method for identifying nucleic acids regulating metastasation | |
| HK1193429A (en) | Treatment of angiogenesis disorders | |
| HK1193429B (en) | Treatment of angiogenesis disorders |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| 121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 24725852 Country of ref document: EP Kind code of ref document: A1 |