NL2028505B1 - Sugar thiol compounds and methods of producing the same - Google Patents

Sugar thiol compounds and methods of producing the same Download PDF

Info

Publication number
NL2028505B1
NL2028505B1 NL2028505A NL2028505A NL2028505B1 NL 2028505 B1 NL2028505 B1 NL 2028505B1 NL 2028505 A NL2028505 A NL 2028505A NL 2028505 A NL2028505 A NL 2028505A NL 2028505 B1 NL2028505 B1 NL 2028505B1
Authority
NL
Netherlands
Prior art keywords
group
compound
formula
alkyl
prot
Prior art date
Application number
NL2028505A
Other languages
Dutch (nl)
Inventor
Hajjaj Bouchra
Herman Veeneman Gerrit
Original Assignee
Pharmacytics B V
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 Pharmacytics B V filed Critical Pharmacytics B V
Priority to NL2028505A priority Critical patent/NL2028505B1/en
Application granted granted Critical
Publication of NL2028505B1 publication Critical patent/NL2028505B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/02Acyclic radicals, not substituted by cyclic structures
    • C07H15/04Acyclic radicals, not substituted by cyclic structures attached to an oxygen atom of the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H13/00Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids
    • C07H13/12Compounds containing saccharide radicals esterified by carbonic acid or derivatives thereof, or by organic acids, e.g. phosphonic acids by acids having the group -X-C(=X)-X-, or halides thereof, in which each X means nitrogen, oxygen, sulfur, selenium or tellurium, e.g. carbonic acid, carbamic acid

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to a sugar thiol compound, a method for preparing said compound, the use of said compound as a promoiety attached to a drug molecule to provide a prodrug, in order to improve oral bioavailability of said drug molecule, to the use of said compound in the preparation of a prodrug, said prodrug and the use thereof. The invention is in the field of medicinal chemistry.

Description

TITLE Sugar thiol compounds and methods of producing the same
TECHNICAL FIELD The present invention relates to a sugar thiol compound, a method for preparing said compound, the use of said compound as a promoiety attached to a drug molecule to provide a prodrug, in order to improve oral bioavailability of said drug molecule, to the use of said compound in the preparation of a prodrug, said prodrug and the use thereof. The invention is in the field of synthetic chemistry.
BACKGROUND The present invention is related to so-called sugar thiol compounds. Sugar 6-thiols are usually prepared by substituting a 6-chloride, a 6-chloride or 8-O-mesyl with sodium thioacetate followed by removal of the S-acetate. Sugars having a 1-O-linker-SH can be made by different methods, depending on the linker moiety.
Sugar thiols are compounds that comprise a sugar group as well as a thiol group. They may be used for several applications, such as the synthesis of dendrimers, carbohydrate cluster molecules, recognition sites in DNA intercalators, and as promoiety for prodrugs.
Prodrugs are often used to enhance drug oral bioavailability of drugs that have a low bioavailability [see the publication: Prodrugs and Targeted Delivery, Rautio, J, (Ed.), 2011, Wiley-VCH, Weinheim, Germany]. Prodrugs, which are inactive or less active derivatives of drug molecules and which undergo enzymatic or chemical transformation within the patient's body to regenerate the active form of said drug, have been a major strategy in meeting this challenge. Different types of prodrugs exist, such as carrier prodrugs that contain a promoiety, i.e. a covalently bound molecule that can e.g. increase the solubility and/or membrane permeability of the drug resulting in a higher oral bioavailability. A special subset of carrier prodrugs are so-called drug-glycosides, in which a sugar part acts as the promoiety. In these prodrugs, the anomeric hydroxyl group of a sugar moiety is covalently linked to a drug molecule, either in a direct manner (e.qg., to a hydroxyl of a drug), or in an indirect manner by use of a linker moiety that can be attached to any functional group in a drug.
SUMMARY It is an object of the present invention to provide new sugar thiol compounds and methods for preparing these.
It is a further object of the present invention to provide a novel sugar thiol compound that can be used as a promoiety for preparing a prodrug of a drug molecule to increase the oral bioavailability of said drug molecule.
In a first aspect, the invention relates to a compound of Formula | according to claim 1. In a second aspect, the invention relates to a method of preparing a compound of Formula | according to claim 11. In another aspect, the invention relates to a use of the compound of Formula | as a promoiety attached to a drug molecule to provide a prodrug, in order to improve oral bioavailability of said drug molecule. In addition, the invention relates to the use of a compound of Formula | in the preparation of compound of Formula lla or IIb according to claim 14. The invention also relates to a compound of Formula lla or IIb and to the use thereof as a medicament, therapy, imaging agent or diagnostic agent.
DETAILED DESCRIPTION The present invention will be disclosed in more detail below.
BRIEF DESCRIPTION OF DRAWINGS Figures 1-1 to 1-9 show table 2 which show compounds according to the present invention.
LIST OF DEFINITIONS The following definitions are used in the present description and claims to define the stated subject matter. Other terms not cited below are meant to have the generally accepted meaning in the field.
“Alkyl” as used in the present description refers to an alkyl group that can be branched or unbranched. Examples of alkyl include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl. 2- methylpentyl, 3-methylpentyl, 2,3-dimethylbutyl and 2,2-dimethylbutyl. C1-C6 alkyl as used in the present description refers to an alkyl group that has one to six carbon atoms in total.
“(hetero)alkyl” as used in the present description refers to an alkyl group (defined above) or a heteroalkyl group (defined below). C1-C6 (hetero)alkyl as used in the present description refers to a (hetero)alkyl group that has one to six carbon atoms in total.
“heteroalkyl” as used in the present description refers to an alkyl group that can be branched or unbranched and that comprises at least one heteroatom, such as O, S, N, F, preferably oxygen (QO), preferably one oxygen, more preferably said heteroalkyl is an alkoxyalkyl group. Examples of heteroalkyl include -CH,CH,OCHj3;, -CH2O0CHsCHa,
and -CH>CH>CH>OCH2CHs. C1-C6 heteroalkyl as used in the present description refers to a heteroalkyl group that has one to six carbon atoms in total.
“Drug” of “drug molecule” as used in the present description refers to a pharmaceutically active agent. This can be an approved drug of medicament, or a candidate drug undergoing laboratory testing, or preclinical or clinical trials.
“Drug moiety” as used in the present description refers to a drug molecule that is part of/attached to the compounds according to present invention.
“Sugar” as used in the present description refers to alpha- and beta-linked monosaccharides, disaccharides, trisaccharides and tetrasaccharides. Mono- saccharides have the general molecular formula (CH20)., where n can be 4, 5 or 6. They can be classified according to the number of carbon atoms in a molecule. Monosaccharides where n is 4 are referred to as tetroses, where nis 5, these are referred to as pentoses, e.g., ribose and deoxyribose, and where n is 8, these are referred to as hexoses, e.g., mannose, glucose and galactose. Disaccharides are made up of two monosaccharide units. Examples of relevant disaccharides are maltose, isomaltose, cellobiose, gentiobiose and lactose. Tri- and tetrasaccharides are oligosaccharides composed of three and four monosaccharide units, respectively. Examples of trisaccharides are maltotriose, isomaltotriose, negerotriose and melezitose. Examples of tetrasaccharides are maltotetraose and nigerotetraose.
“Oral bioavailability” as used in the present description refers to the extent and rate at which a drug enters the systemic circulation after oral administration, thereby becoming available to access the site of desired action. Oral bioavailability in the context of the present invention is herein defined as the fraction of an orally administered drug that reaches the systemic circulation.
“Prot” or “protective group” as used in the present description refer to groups that are a reversibly formed derivative of an existing functional group in a molecule, in this case the -OH group. The protective group is temporarily attached to decrease reactivity of the -OH group so that the protected functional group does not react under synthetic conditions to which the molecule is subjected in one or more subsequent steps.
In the present invention, compounds having one or more of these protective groups are precursor compounds and in case the compound according to the invention is e.g. for a further synthesis, e.g. as a promoiety for a prodrug, these one or more protective groups need to be removed for the final compound obtained (e.g. the prodrug) to be active. However, this removal of one or more of these protective groups can be done in the sugar-thiol stage or after the compound has been formed into the final compound {e.g. the prodrug).
DESCRIPTION OF EMBODIMENTS In a first aspect, the present invention relates to a compound according to Formula | as disclosed in claim 1. R1 R2b 0 \ O—R5 R2a / R3 R4 Formula | In this Formula |, the groups are as discussed below.
R1 groups R1 is selected from the group consisting of -H, -F, -OH, -O-Prot, and -SH.
In case R1 is -SH, then R5 cannot comprise a thiol (SH) group, since the compounds according to the invention only have one thiol group.
R2 groups R2a and R2b are each independently selected from the group consisting of -H, - OH, -O-Prot and -F; with the provision that one of R2a and R2b is -H and the other of R2a and R2b is selected from the group consisting of -OH, -O-Prot and -F.
In other words, R2a and R2b cannot both be the same, one of them is always -H and the other is either -OH, the protective form thereof {(-O-Prot) or -F.
R3 groups R3 is -OH or the protective form thereof (-O-Prot). R4 groups R4 is selected from the group consisting of -OH, the protective form thereof (-O- Prot) or -F.
R5 groups R5 is selected from the group consisting of -WSH, -CH2CH2O0CH:CH:SH, - (C=0O)N(RB)YSH, -(C=0)-4-piperidine-SH, -(C=0)-3-pyrrolidine-SH, -(C=0)-3-azetidine- SH, C1-C6 (hetero)alkyl, and -(C=0)NR7RS.
In these options for R5, the following is observed: * R6 is hydrogen or a C1-C6 (hetero)alkyl, preferably selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and -CH2CHz2OCH;3;
* Wis a C1-C6 alkyl group, preferably -CH2CH:- or -CH2CH2CHz-; W is a linker group that couples the sugar ring to the thiol group; *Y is a C1-C6 (hetero)alkyl, preferably selected from the group consisting of -CH:-,- CH2CH:-, and -CH2CH2CHs-; Y is a linker group coupling the amide group ((C=0)N(R8&)) 5 to the thiol group; * R7 is hydrogen or a C1-C6 alkyl, preferably selected from the group consisting of hydrogen, methyl, ethyl, and n-propyl; * R8 is a C1-C6 alkyl, preferably selected from the group consisting of methyl, ethyl, and n-propyl; In case a thiol group (SH) is present in R5, then R1 cannot comprise a thiol (SH) group, since the compounds according to the invention only have one thiol group. Provisos regarding the thiol group The compound of the invention has the following provisos since the compounds according to the invention only have one thiol group: * when R1 is -SH, then R5 is a C1-C8 (hetero)alkyl or -(C=O)NR7R8; * when R5 is selected from the group consisting of -YSH, -CH2CH20CH2CH2SH, - (C=O)N(R6)WSH, -(C=0)-4-piperidine-SH, -(C=0)-3-pyrrolidine-SH, and -(C=0)-3- azetidine-SH, then R1 is selected from the group consisting of -H, -F, -OH, and -O-Prot.
In an embodiment, each Prot group is independently selected from the group consisting of acetyl, benzoyl, benzyl, 4-methoxybenzyl, trialkylsilyl, most preferably acetyl. If there are more than one Prot groups present in the compound, they can either be all the same or they can be more than two of the list above. In a preferred embodiment, all of the R1, R2a or R2b, R3 and R4 groups (if not -F) are either -OH or -O-Prot, so in this embodiment all hydroxyl groups that are present in the compound are either not protected (so -OH) or are all protected (so -O-Prot), specifically preferred if they are all - O-Prot, they are all acetyl. The removal of said protective groups can be carried out by any means known to a skilled person.
The table below shows the structures of these compounds according to the invention.
JH o u o i SH ú Ts TT : o_ ‚0 HON Op Ho N00 HO Foy OH HO "OH HO OH
OH OH OH SH SH ! ’ 0 LT o NT Og NSH o 7 J Q_ 0 “LL HO 0 Qo HO eg HO I “OR Len HO “OH
A OH OH > ? J | : Og NSH Oy NSH OM SH HO SOO o_.0 00 JJ Hoy LNG HO ‘OH voro HOY YF
OH OH SH ' HS Q Po \ / Yo DN ONO gg oN H HO AN o o_,0 /
HON BE Hs °° HO OH HO “OH OH HO “OH
OH HS o / A Hy _ , ot oO Nn Noo Nett” o ~ 0 8 0 & eH A ; A -. . HOw ed A gg Age TL A HO 90 oo HG OH Ho OH od Op OR 0 Oy NSH HO 0_,0 HO” “OH
OH The present inventors have also found a novel way of producing the novel sugar thiols according to the present invention. The method comprising:
R1' AG iii RS O-R5' — + Formulal R2a" R3 R4 Formula Ib Scheme 1: General synthesis of sugar thiols wherein R1' is selected from the group consisting of -H, -F, -OH, -O-Prot, and X; R2a, R2b, R3 and R4 have the same meaning as described for Formula |; R5' is selected from the group consisting of -W-X, -CH:sCH2OCH:CH:X, - (C=O)N(R6)YX, -(C=0)-4-piperidine-X, -{C=0)-3-pyrrolidine-X, -(C=0)-3-azetidine-X, C1-C6 (hetero)alkyl, and -(C=O)NR7R8; X is -OS(=O}2CHs or a halide selected from the group consisting of chlorine, bromine, and iodine; R6 is hydrogen or a C1-C6 (heterojalkyl, preferably selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, and -CH2CH2OC Ha; W is a C1-C8 alkyl group, preferably -CH2CH:- or -CH2CH:CH:-; W is a linker group that couples the sugar ring to a halide or methane sulfonate group; Y is a C1-C6 (hetero)alkyl, preferably selected from the group consisting of -CH:- ,-CH2CH3-, and -CH2CH2CH.-; Y is a linker group coupling the amide group ((C=0)N(R86)) to the halide group; R7 is hydrogen or a C1-C8 alkyl, preferably selected from the group consisting of hydrogen, methyl, ethyl, and n-propyl, R8 is a C1-C6 alkyl, preferably selected from the group consisting of methyl, ethyl, and n-propyl; with the provision that: in case R1’ is X, then R5’ is C1-C6 (hetero)alkyl or -(C=O)NR7R8; in case a halide (chlorine, bromine, iodine) or methane sulfonate group (X) is present in RS’, then R1' cannot comprise a halide or methane sulfonate (X) group, since the intermediate compounds used to prepare the compounds according to Formula | only have one halide or methane sulfonate group, or in other words, in case R5’ is selected from the group consisting of -W-X, -CH2CH2OCH:CH:X, -(C=O)N(R6)YX, -(C=0)-4-
piperidine-X, -(C=0)-3-pyrrolidine-X, and -(C=0)-3-azetidine-X, then R1’ is selected from the group consisting of -H, -F, -OH, and -O-Prot; wherein the method comprises subsequent steps i) and ii): step i) comprising reacting a compound of Formula Ib with potassium thioacetate to obtain a thioacetate-adduct of a compound Formula Ib; and step ii} comprising reacting the thioacetate-adduct of the compound Formula Ib obtained in step i) with sodium methoxide to obtain the compound according to Formula |.
In a specific embodiment of the compound or method of the invention, in the compound of Formula |, at least one of R1, R2a, R2b or R4 is -F. So at least one fluorine substituent is present in the thiol compound.
In a specific embodiment of the compound or method of the invention, in the compound of Formula |, when R5 is -WSH in which W is ethyl or propyl (a C2- or C3-alkyl group) [in other words R5 is -CH2CH:SH or -CH:2CH2CH2SH], then: i) R1 is -H or -F; and/or ii) R2a and R2b are each independently -H or -F; with the provision that one of R2a and R2b is -H and the other of R2a and R2b is -F [R2a and R2b cannot be the same]; and/or iii) R4 is -F. In this specific embodiment, when R5 is -CH2CH2SH or - CH2CH2CH:SH, R1, R2, R3 and R4 cannot all be/comprise an -OH or -O-Prot group, in other words the ring structure cannot be an unsubstituted galactopyranose or glucopyranose ring.
Reaction step i} comprising the step of reacting a compound of Formula Ib comprising a halide or methane sulfonate with potassium thioacetate, is carried out in a solvent or a mixture of solvents, preferably selected from the group consisting of DMF, DMA or NMP. This reaction step i) is preferably carried out at a temperature of between 40 and 90 °C, such as between 50 and 80 °C. In a specific embodiment it is carried out at 65 °C. Reaction step i) is preferably carried out under an inert atmosphere, e.g. in the absence of oxygen, preferably under an argon or nitrogen atmosphere. Reaction step i) is carried out for a duration of 0.5 to 24 hours.
Reaction ii) step comprising the step of reacting the thioacetate-adduct of the compound Formula Ib obtained in step i) with sodium methoxide, potassium carbonate or sodium methanethiolate, is preferably carried out in methanol or in a mixture of methanol with other solvents, such as tetrahydrofuran of 1,4-dioxane. This reaction step ii) is preferably carried out at a temperature of between 15 and 30 °C. In a specific embodiment it is carried out at 20-25 °C. Reaction step ii) is preferably carried out under an inert atmosphere, e.g. in the absence of oxygen, preferably under an argon or nitrogen atmosphere. Reaction step ii) is carried out for a duration of 1 to 6 hours.
The method as described above may further comprise a step 0), prior to step i), said step o) comprising reacting a compound according to Formula III R1'— R2b ‚70 nd A R2a” \ R3 R4 Formula Il! with a bromo alcohol of formula Br-{C1-C8)-alkyl-OH, to provide a compound according to Formula Ib wherein R5’ -W-X, in which W is (C1-C6)-alkyl, and in which X is Br. A specific embodiment of the method is shown below. This is according to claim 11, in which W is alkylene and n = 1-5.
‚Ri 1 Rt R2b I R2b a Ref [ La 19 . Br yy PN SAc RIN ORS re” No TM RoT R4 R4 R4 - R1 R2b I R2a 1 ; r 3SH Ra” opt a R4 In another aspect, the present invention relates to the use of a compound according to Formula | as a promoiety attached to a drug molecule to provide a prodrug, in order to improve oral bioavailability of said drug molecule. As disclosed below in the examples, the use of these compounds as promoiety in a prodrug significantly increases the oral bioavailability of said drug compound. In another aspect, the present invention relates to the use of a compound of Formula | in the preparation of a compound of Formula lla or lb or pharmaceutical acceptable salts thereof
DM[Z]—L—S R1 \ O-R5 3 O—V—L—[Z]DM R23 / R2a 4 R3 R4 R3 R4 Formula lia Formula lib In these prodrug compounds R2a, R2b, R3, R4, R6, R7, R8, W, and Y are disclosed above. In addition, for R1 (Formula IIb} all groups except the thiol groups are possible, so that R1 is selected from the group consisting of -H, -F, -OH, and -O-Prot. In addition, for RS {Formula lla) all groups except the thiol groups are possible, so that R5 is a C1-C8 (hetero)alkyl or -(C=O)NR7R8.
In Formula llb V is selected from the group consisting of -WS-, - CH2CH>OCH2CH:S-, -(C=0)N(R8)YS-, -(C=0)-4-piperidine-S-, -(C=0)-3-pyrrolidine-S-, and -(C=0)-3-azetidine-S-; V is a thiol-containing linker group linking the sugar moiety to the drug moiety (via linker L).
In Formula lla and Formula IIb L is a linker molecule linking the drug moiety to the thiol group of the sugar thiol compound (according to Formula |). L may be any suitable linker molecule but is preferably -S-CH(R9)-O-C(=0). RS is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, and t- butyl, preferably hydrogen or methyl. DM is a drug moiety and [Z] represents a part of DM; and Z is selected from the group consisting of O, S, and N.
In an aspect, the invention relates to a compound according to Formula lla or IIb and in another aspect, to a compound according to Formula lla or IIb for use as a medicament, therapy, imaging agent or diagnostic agent.
In an embodiment, each Prot group is independently selected from the group consisting of acetyl, benzoyl, benzyl, 4-methoxybenzyl, trialkylsilyl, most preferably acetyl. If there are more than one Prot groups present in the compound, they can either be all the same or they can be more than two of the list above. In a preferred embodiment, all of the R1, R2a or R2b, R3 and R4 groups (if not -F) are either -OH or -O-Prot, so in this embodiment all hydroxyl groups that are present in the compound are either not protected (so -OH) or are all protected (so -O-Prot), specifically preferred if they are all - O-Prot, they are all acetyl. The removal of said protective groups can be carried out by any means known to a skilled person.
The linker of the promoiety is covalently bound to the functional group ZH of a drug molecule (DM-ZH) representing small molecules to therapeutic peptides, to provide the compounds according to the present invention. ZH is part of the drug molecule and represents an alcohol, phenol, oxime, a primary or secondary amine or a thiol, with the provision that NH and NH: are not part of an amide, carbamate or urethane within said drug molecule. “DM” denotes a drug moiety which forms with one of its OH, NHz, NH or SH functional group, the active drug; it is to be understood that in an embodiment, the carbonyl moiety of the 1-substituted-(disulfanyl)alkyl is linked to the OH, NH2/NH or SH group of the active drug to form a carbonate-, carbamate- or thiocarbonate-type linkage. The drug moieties with its attached OH, SH, NH: or NH functional groups preferably have a molecular weight in the range of 100-1000 daltons. The invention can be applied to many drugs but may be applied especially to drugs that have one or more imperfections, such as poor solubility, permeability, (oral) bioavailability or dissolution rate, or the induction of gastrointestinal side effects, undesired metabolism or bad taste. In those cases, it is preferred that G is selected from a structural motif that optimizes at least one of these drug imperfections.
Representative amine-containing drugs that can be used in the compounds according to the present invention include 5'-Deoxy-5-fluorocytidine, Cytarabine, Lenalidomide, Thalidomide, Acyclovir, Doxorubicin, Losartan, Orciprenaline, Albendazole, Duloxetine, Mesalazine, Linagliptin, Atomoxetine, 5-Fluorouracil, Methylphenidate, Palbociclib, Azacitidine, Gabapentin, Metoprolol, Nintedanib, Carvedilol, Gemcitabine, Rasagiline, Pscilocin, Celecoxib, Ibrutinib, Riluzole, Meropenem, Cinacalcet, Lapatinib, Tamiflu, and Ceftriaxon. Representative hydroxy- containing drugs that can be used in the compounds according to the present invention include Abiraterone, Fesoterodine, Rotigotine, Ciclopirox, Acyclovir, Fulvestrant, Tenofovir, Azacitidine, Ganciclovir, Testosterone, Cytarabine, Kalydeco, Tizoxanide, Cannabidiol, Paliperidone, Venlafaxine, Edaravone, Paracetamol, Vorinostat, Gemcitabine, Paclitaxel, Pscilocin, Estradiol, Propofol, and Orciprenaline. Representative thiol-containing drugs that can be used in the compounds according to the present invention include mercaptopurin, acetylcysteine, bucillamine, captopril, and zofenoprilat. In an embodiment, the drug DM-ZH is selected from the group consisting of Abiraterone, Cinacalcet, Duloxetine, Ritalin and Mercaptopurin.
The present invention has as one of its aims to increase the oral bioavailability of drugs. Oral bioavailability is usually assessed by determining the area under the plasma concentration—time curve (AUC) [see publication ADMET for medicinal chemists, Tsaioun, K and Kates, S.A. (Eds.), 2011, Ch. 5, Wiley]. Plasma drug concentration increases with extent of absorption, the peak concentration is reached when drug elimination rate equals absorption rate. Peak time is the most widely used general index of absorption rate; the slower the absorption, the later the peak time. The most reliable measure of a drug's oral bioavailability is AUC. The AUC is directly proportional to the total amount of unchanged drug that reaches systemic circulation. Drug products may be considered bicequivalent in extent and rate of absorption if their plasma concentration curves are essentially superimposable. In practical terms, the oral bioavailability is the percentage of the AUC of a drug available in the blood of a test species after oral administration in relation to the AUC obtained from the same dose administered intravenously to the test subject. A broad spectrum of methods is available for determining intestinal absorption of compounds in experimental animals. Typical laboratory methods include perfusion via (multiple) lumen tubes, mass balance studies and blood kinetics following oral and intravenous administration of the compound [see http://www .rivm.nl/bibliotheek/rapporten/630030001.pdf]. Relevant animal species include mice, rats, dogs, mini pigs and monkey. Oral bioavailability of a drug and its conjugate can also be predicted to some extent using appropriate in vitro models [see publication Altern. Lab. Anim., 2001, 29, 649-668]. Appropriate in vitro tissue models include everted gut sac, perfused intestinal segments and Ussing chambers. Cell-based in vitro models include small-intestinal cell lines from fetal and neonatal rats and Caco-2 cells.
The term “increasing the oral bioavailability of a drug” or “increased bioavailability” is used herein to indicate that the oral bioavailability of a drug modified according to the invention is increased in comparison to the unmodified drug (being DM- ZH]. Even a small increase of oral bioavailability can be relevant. For instance, if the drug currently has an oral bioavailability of 10%, an increase to 11 or 12% using the compounds according to the invention is considered a relevant increase. For example, a drug currently having an oral bioavailability of 10% may form a prodrug of the invention which, upon oral administration, leads to the accumulation of the unconjugated drug with an oral bioavailability of more than 10%. The increase in oral bioavailability may be in the order of a few percent points, resulting in an increased bioavailability of 11%, 12%, 13%, 14% or 15% or even more, such as up to 20%. More spectacular increases have also been observed; depending on the drug and type of prodrug, oral bioavailability of up to 30%, 40% or 50% or even more such as 60% up to 70% appeared achievable. In certain cases, the increase was even more, such as 71 up to 90%. In exceptional cases, even higher oral bioavailability may be achieved such as 91% up to even 100%.
In an embodiment, the present invention relates to the use of the compound as a linker attached to a drug molecule to improve the oral bioavailability by at least 1 %, preferably by at least 2 % compared to the oral bioavailability of the drug molecule as such. With an increase of a certain % an incremental increase is meant. When the oral bioavailability of a certain drug is 10 % and after it has been prepared in a compound according to the invention the oral bioavailability of said drug is 11 %, this is considered an increase of 1 %. The increase of oral bioavailability achieved by the method according to the invention may depend on the drug and type of prodrug used. It has been observed that adrug conjugate as prepared using a method according to the invention leads to a higher concentration of the drug (i.e., without the conjugated promoiety) in circulation upon oral administration, compared to the concentration of the same unconjugated drug when administered orally. Other variations to the disclosed embodiments can be understood and effected by those skilled in the art in practicing the claimed invention, from a study of the drawings, the disclosure, and the appended claims. In the claims, the word "comprising" does not exclude other elements or steps, and the indefinite article "a" or "an" does not exclude a plurality. The scope of the present invention is defined by the appended claims. One or more of the objects of the invention are achieved by the appended claims.
EXAMPLES The present invention is further elucidated based on the Examples below which are illustrative only and not considered limiting to the present invention. LC-MS data were recorded on an Agilent 1200 Infinity UPLC system, attached to an Agilent 6100 single quadrupole MS detector. A Kinetex 2.6py EVO C18 100A column of 50x2.1 mm equipped with a EVO C18 guard column (Phenomenex) was used. The LC- MS experiments were run at a flow speed of 0.6 mL/min with a weakly acidic solvent system consisting of 0.1% formic acid in water (A) and acetonitrile containing 0.1% formic acid (B) was used. A gradient was run from 5% B to 60% B in 1.0 minutes, followed by a gradient from 60% to 95% B in 2.0 minutes and keeping the gradient at 95% B for 1 to 6 minutes. SYNTHESIS OF COMPOUNDS ACCORDING TO FORMULA | The below synthesis shows the formation of a specific galactose O-linked thiol compound 12 according to Formula I.
OAc OAc OAc AcO oe a AcO ke a AcO e. ACO done TTT Aco No TTT Aco oe OAc OAc OAc 10 11 | lla
OH HO" ® o— SH
OH 12 Synthesis of intermediate compound 10 Dry penta-O-acetyl-B-D-galactopyranoside (5.0 g, 13 mmol, 1.0 eq.) was dissolved in anhydrous DCM (30 mL) and cooled to 0 °C under an inert atmosphere of nitrogen. 2-bromoethanol (1.8 mL, 26 mmol, 2.0 eq.) was added, followed by the dropwise addition of boron trifluoride diethyletherate (4.7 mL, 38 mmol, 3.0 eq.). The reaction was warmed up to room temperature. After 1h at ambient temperature, the reaction mixture was diluted with EtOAc, then washed with a saturated solution of NaHCO; saturated and brine. The organic layer was dried with MgSOs and concentrated under vacuum. The crude mixture was purified by flash chromatography (silica, 10 to 70% EtOAc in heptane) to afford the desired compound 10 as a transparent oil that crystallised over time (4.25 g, 9.34 mmol, 73%). Synthesis of intermediate compound 11 To a solution of the bromide 10 (4.25 g, 9.34 mmol, 1.0 eq.) in DMF (12 mL) under an inert atmosphere of nitrogen was added potassium thioacetate (1.6 g, 14 mmol.
1.5 eq.). The reaction mixture was stirred under Ny at 80 °C until full conversion. The crude mixture was dissolved in EtOAc (100 mL), washed with brine, a 2 M solution of NaOH and brine. The organic layer was dried over MgSO., filtered and evaporated to dryness. The crude mixture was purified by flash chromatography (silica, 5 to 70% EtOAc in heptane) to afford the desired product 11 as a pale orange oil (3.73 g, 8.28 mmol, 88%).
Synthesis of compound 12 according to Formula | To a solution of 11 (700 mg, 1.54 mmol, 1eq.) in MeOH (10 mL), NaOMe (84mg,
1.54mmol, 1eq.) was added. The reaction was stirred under an inert atmosphere of nitrogen for 6 h. The reaction mixture was neutralized with Dowex® (50Wx8 100-200 mesh), filtered and concentrated in vacuo yielding 12 as a clear oil in a quantitative yield. Using the same method as discussed above for compound 12, the formation of a specific glucose O-linked thiol compound 13 can be carried out.
OH
H HO” o~SH
OH 13 Synthesis of compound 13 according to Formula | Compound 13 was obtained in a similar manner to that described for compound 12 starting with penta-O-acetyl-B-D-glucopyranoside. kkk The below synthesis shows the formation of a specific glucose O-linked thiol compound 17 according to Formula I.
Cl B 1 cl r s 9 HN A, 0 ’ ° A o o 0 o o 0,5 J Hb J TE eye 00; Fv “0 __ Fx “0 _ J , Ô ae o NY iede & & & 14 15 16 -
J FOH
OH 17 Synthesis of intermediate compound 15 To a solution of 2,3,6-tri-O-benzoyl-4-fluoro-4-deoxy-D-glucopyranosyl trichloro acetimidate 14 (disclosed in WO2010/77623) (4.19, 6.4, 1eq.) in DCM (20.0 mL) containing 4A MS was added 2-bromoethanol (2.4 g, 19.3 mmol, 3eq.) and the solution was cooled to 0 °C. To this solution was added BF; O(C2Hs)2 (0.95 mL, 7.7 mmol, 1.2eq.) and the resulting solution stirred at 0°C for 2 h. After this time Et:N was added and the solution filtered, concentrated and purified by flash chromatography to give 15 (2.32 g,
3.9 mmol, 60%). Synthesis of intermediate compound 16 Compound 16 was obtained in a similar manner to that described for compound 11 starting with 15. Yield = quantitative Synthesis of compound 17 according to Formula | Compound 17 was obtained in a similar manner to that described for compound 12 starting with 16. Yield = quantitative.
The below synthesis shows the formation of a specific O-linked thiol compound 23 according to Formula |.
OH Aon B20 AcO ej + ue EE ode Lx He Ve Ve oyhe : B20 y 98e OH Ô Sas 0 AcO oe 0 oH 18 1 2 HO AcO AcO rob : Vis oyhe : o ve wordy : 0 HO” ~"0" "SH ACO” os Aco oo OH OAc OAc 23 22 2 Synthesis of intermediate compound 18 To a solution of (3R)-butane-1,3-diol (2.25 g, 25 mmol, 1 eq.) in pyridine (14 mL) stirred at 0°C was slowly added TBDMS-CI (7.1 mL, 27.5 mmol, 1.1 eq.) followed by DMAP (31 mg, 0.25 mmol, 0.1 eq.). The reaction mixture was stirred for 18h at room temperature under an inert atmosphere of nitrogen. The reaction mixture was diluted with water and extracted several times with DCM. The organic layers were combined, dried over MgSO, and concentrated yielding compound 18 (6.8 g, 20.6 mmol, 82%).
Synthesis of intermediate compound 19 To a solution of isopropyl 2,3,4,6-tetra-O-benzoyl-B-D-1-thiogalactopyranose (17.5 g, 27 mmol, 1.3 eq.), compound 18 (6.8 g, 21 mmol, 1 eq.) and molecular sieves (4 A) in DCM (104 mL) was added NIS (7 g, 31 mmol, 1.5 eq.) and triflic acid (183 mL, 21 mmol, 0.1 eq.). The solution was stirred at room temperature under an inert atmosphere of nitrogen for a few minutes. The reaction mixture was diluted with DCM and washed with a solution of sodium thiosulfate and a saturated solution of NaHCO:. The organic layer was dried over MgSO,, filtered, evaporated to dryness and purified by column chromatography yielding 19 (13.9 g, 15.3 mmol, 74%).
Synthesis of intermediate compound 20 Step lll-c): To a solution of compound 19 (13.8 g, 15.2 mmol, 1 eq.) in MeOH (40 mL) and dioxane (40 mL) was added NaOMe (1.6 g, 30 mmol, 2 eq.) and the resulting solution was stirred at room temperature until completion. The reaction mixture was neutralized with Dowex H*, filtered and concentrated. The residue was coevaporated with pyridine and used as such in the next step.
Step IV-c): To the material obtained in the previous step (15.2mmol, 1eq.) in pyridine (80 mL) was added acetic anhydride (8.6 mL, 91.2 mmol, 6 eq.). The reaction mixture was stirred overnight at room temperature under an inert atmosphere of nitrogen. The solution was concentrated and coevaporated with toluene. The residue was dissolved in EtOAc and washed with a 1M solution of HCI, water, a saturated solution NaHCO; and brine. The organic layers were dried over MgSO,4, filtered and purified by flash chromatography (silica, 0 to 50% EtOAc in heptane) yielding the desired product (7.29, 11 mmol, 72%).
Step V-c): To the material obtained in the previous step (7.2 g, 11 mmol, 1 eq.) in THF (75 mL) was added acetic acid (627 pL, 11 mmol, 1eq.) and a 1M solution of TBAF in THF (11 mL, 11 mmol, 1 eq.). The reaction mixture was stirred at room temperature for 24h. The solution was then concentrated and purified by flash chromatography (silica, to 100% EtOAc in heptane) to give compound 20 (3.7g, 8.9mmol, 81%). Synthesis of intermediate compound 21 To a solution of compound 20 (3.4 g, 8.1 mmol, 1 eq.) in pyridine (65 mL) was added MsCI (1.3 mL, 16.3 mmol, 2 eq.) at 4°C under an inert atmosphere of nitrogen. 20 The reaction mixture was then stirred at room temperature under Ns for 1h. The mixture was concentrated, dissolved in EtOAc and filtered. The filtrate was washed with a 0.5 M solution of HCI, water, and a saturated solution of NaHCO:. The organic layer was dried over MgSO., filtered and evaporated to dryness yielding compound 21 (3.6 g, 7.2 mmol, 89%). LC-MS (ESI): r.t. = 2.64 min, m/z calcd. for C1sH30013S = 498.1; found m/z = 521.2 [M+Na]".
Synthesis of intermediate compound 22 To a solution of the compound 21 (3.6 g, 7.2 mmol, 1 eq.) in DMF (15mL), potassium thioacetate (2.6 g, 22.4 mmol, 3.1 eq.) was added. The reaction mixture was stirred at 50°C for 1h under an inert atmosphere of nitrogen. The reaction mixture was the diluted with EtOAc and washed with water, and brine. The organic layer was dried over MgSO., filtered, concentrated in vacuo and purified by column chromatography (silica, O to 60% EtOAc in heptane) yielding compound 22 (1.9 g, 4.0 mmol, 56%) as a red oil.
Synthesis of compound 23 according to Formula | To a solution of 22 (574 mg, 1.2 mmol, 1 eq.)in MeOH (10mL) was added NaOMe (130 mg, 2.4 mmol, 2 eq.) and the resulting solution stirred at room temperature under an inert atmosphere of nitrogen until completion.
The reaction mixture was neutralized with Dowex H*, filtered and concentrated to give 23 (320 mg, 1.2 mmol, 99%). LC-MS (ESI): r.t. = 0.64 min, m/z calcd. for C1pH2006S= 268.1; found m/z = 291.0 [M+Na]*, m/z = 267.0 [M-H] and m/z = 313.0 [M-H+HCOOH]. The below synthesis shows the formation of a specific O-linked thiol compound 28 according to Formula I. i” cl 5 OH wor + { ks oy ’ — oy ’ AcO Tr OAc A _oH AcO Tr OAc AcO Tr OAc 24 25 5 x x 0:50 HON O° Aco 0,0 ol o Is LT, H ool J OAc Aco LJ OH OAc AcO OAc OAc 28 27 26 Synthesis of intermediate compound 24 To a solution of dry 2,3,4,6-tetra-O-acetyl-D-galactopyranosyl trichloroacetimidate (4.4 g, 8.9 mmol, 1eq.) and (R}-2-(benzyloxy)-propan-1-ol (1.5 g, 8.9 mmol, 1 eq.) in DCM (20 mL) was added BF3:O(C:Hs): (1.6 mL, 13.4 mmol, 1.5 eq.) at - 10 °C under an inert atmosphere of nitrogen.
The reaction mixture was then allowed to slowly warm up to room temperature.
After 2h, TEA (2.10 mL, 15.1 mmol, 1.7 eq.) was added and the solution was filtered.
DCM (50 mL) was added to the filtrate.
The mixture was washed with a saturated solution of NaHCO: and brine, dried over MgSO. and concentrated.
The crude product was purified by flash chromatography (silica, O to 50% EtOAc in heptane) yielding 21 (2.5 g, 5.1 mmol, 57%) as a slightly yellow oil.
LC-MS (ESI): r.t. = 2.99 min, m/z calcd. for C,,H,,0,, = 496.2; found m/z = 519.2 [M+Na]*.
Synthesis of intermediate compound 25 Compound 24 (2.5 g, 5.1 mmol, 1 eq.) was dissolved in MeOH (50 ml) and 54 mg Pd/C was added.
The reaction was stirred at room temperature under Hz.
Upon completion, the reaction mixture was filtered through celite and concentrate in vacuo yielding 25 (2.0 g, 5 mmol, 98%) as a slightly yellow resin.
The compound 25 was used in the next step without further purification.
Synthesis of intermediate compound 26 The alcohol 25 (1.2 g, 3 mmol, 1 eq.) was dissolved in dry DCM (12 mL) under an inert atmosphere of nitrogen.
TEA (418 uL, 3 mmol, 1 eq.) was added to the reaction mixture followed by mesyl chloride (232 HL, 3 mmol, 1 €q.). The reaction mixture was stirred at room temperature till completion.
The mixture was diluted with DCM, washed with water and brine, dry over MgSQ4 and concentrated to give compound 26 (1.45 g, 3 mmol, quantitative). Compound 26 was used without further purification in the next step.
Synthesis of intermediate compound 27 To a solution of compound 26 (1.45 g, 3 mmol, 1 eq.) in DMF (20 mL) under an inert atmosphere of nitrogen was added potassium thioacetate (2.4 g, 21 mmol, 7 eq.). The reaction mixture was stirred under Nz at 80 °C for 1 hour.
The reaction mixture was then concentrated and dissolved in EtOAc.
The solution was washed with brine, dried over MgSO. and concentrated to dryness.
The crude material was purified by flash chromatography (silica, O to 50% EtOAc in Heptane) to give 27 (950 mg, 2.05 mmol, 68%) as a dark red/brown resin.
Synthesis of compound 28 according to Formula | Deacetylation of compound 28 was performed in a similar manner to that described for compound 23. rk The below synthesis shows the formation of a specific hydroxy intermediate compounds 30a, 30b, 30c or 30d according to Formula I.
NO,
0.0 od AcO + 29a to d Ie + 3atod AcO “OAc OAc | Oy Neon Sy OH Aco o 0
H vol Lone OAc 29a 30a
ON A OH TNT OH AcO 0 0 vol Jon OAc 29b 30b
OH iJ HN, OH Aco 9-0 od on OAc 29C 30c
OH o NT
Y HN] >—OH Aco OO vol Lon OAc 29d 30d Synthesis of intermediate compound 30 To a solution of p-nitrophenyl 2,34 6-tetra-O-acetyl-p-D-galactopyranosyl carbonate (3.9 mmol, 1eq.) (see Bioorg. Med. Chem. Lett., 2016, 26, 3774) and 29a or 29b or 29c or 29d (3.9 mmol, 1 eq.) in DCM (55 mL) was added TEA (7.8 mmol, 2 eq.). The solution was stirred at room temperature under an inert atmosphere of nitrogen until completion. The reaction mixture was diluted with DCM and washed with a 1M solution of HCI and brine. The organic layer was dried over MgSO, filtered and evaporated to dryness. The crude compound was purified by column chromatography yielding the compounds 30a or 30b or 30c or 30d. Compound 30a was obtained in a yield of 94% (2.44 min; m/z = 472.2 [M+Na]+). Compound 30b was obtained in a quantitative yield (2.46 min; m/z = 486.2 [M+Na]*). Compound 30c was obtained in a yield of 86% (2.47 min; m/z = 514.2 [M+K]*). Compound 30d was obtained in a yield of 81% (2.36 min; m/z =470.2 [M+Na]"). The below synthesis shows the formation of specific thioalkyl-linked carbamates 33a, 33b, 33c and 33d. ab Ha Mab 30c-d le od Stad WHE A32a.d VE 33a.
| | | Os Non ON oms OM sac Oy Ns Oo. Oo AcO 0,0 Aco 0.9 HO 0.0 AcO LT wok one wok ome ol H AcO “OAc OAc OAc OH Oe 31a 32a 33a 30a OM 0 H 0 ON She 0 Opi 0_,0 HO AcO “OAc MO pO Oe OAc ¢ 39 33b 31b 30b OH | SAc SH oJ oJ oJ oJ Aco 90 “XT, “XT, el lyf OAc AcO I Oe AcO I Ae HO I OH 30c 31c 32¢ 33C OH | SAc SH 0 NT 0 NT o NT o NT 7 0 3 0 $ 0,0 Aco NX wao eT "rol J 4 , HO "OH ne Ac AcO Te OAc AcO Te OAc OH 33d 30d 31d 32d Synthesis of intermediate compounds 31a or 31b To a solution of 30a or b (3.67 mmol, 1 eq.) in pyridine (16.6 mL) was added mesyl chloride (7.34 mmol, 2 eq.) at 4°C. The reaction mixture was then stirred at room temperature under an inert atmosphere of nitrogen until completion. The rection mixture was concentrated. The residue was dissolved in EtOAc. The suspension was filtered. The filtrate was washed with a 0.5 M solution of HCI, water, and a saturated solution of NaHCO:. The organic layer was dried over MgSO, and evaporated to dryness yielding 31a or b. Compound 31a was obtained as an oil from 30a. Yield = 98%. LCMS (ESI): rt =2.61 min, m/z calcd. for C,H, NO, ,S = 527.13, found m/z = 5650.2 [M+Na]* Compound 31b was obtained as an oil from 30b. Yield = 86%. LCMS (ESI): rt = 2.815 min, m/z calcd. for C,,H,,CINO,, = 481.1, found m/z = 504.2 & 506.2 [M+Na]". Synthesis of intermediate compounds 31c ord To a solution of 30c or 30d (8.5 mmol, 1 eq.) in toluene (85 mL) were added successively imidazole (25.6 mmol, 3 eq.), PPh3 (17.1 mmol, 2 eq.) and 1: (12.8 mmol,
1.5 eq.). The reaction mixture was stirred at 110 °C until completion. The reaction mixture was quenched by adding a saturated solution of NaHCO:. The aqueous layer was extracted with EtOAc. |; was added to the combined organic phases until a persistent brown color was observed. The organic phase was washed with a saturated aqueous solution of Na2S20O3, dried over Na2S0,, filtered and concentrated under reduce pressure. The product was purified by flash chromatography yielding 31¢ or 31d. Compound 31¢ was obtained from 30c. Yield = 74%. LCMS (ESI): rt = 3.02 min, m/z calcd. for C,,H,5INO,, = 585.0, found m/z = 608.2 [M+Na]*. Compound 31d was obtained from 30d. Yield = quantitative. LCMS (ESI): rt = 2.80 min, m/z calcd. for C1sH24INO44 = 557.0, found m/z = 580.0 [M+Na]*. Synthesis of intermediate compounds 32a, 32b, 32c, or 32d To a solution of 31 (a to d) (2.9 mmol, 1eq.) in DMF {8.5 mL} under N» was added potassium thioacetate (8.9 mmol, 3 eq.). The reaction mixture was stirred under N; at 50°C until completion. The reaction mixture was diluted with EtOAc and washed with brine (3 x). The organic layer was dried over MgSO, filtered and evaporated to dryness. The crude compound was purified by column chromatography. Compound 32a was obtained as a brown oil from 31a. Yield = 77%. LCMS (ESI), rt = 2.78 min, m/z calcd. for C,H, NOS = 507.1, found m/z = 530.2 [M+Na]*. Compound 32b was obtained as a brown oil from 31b. Yield = 95%. LCMS (ESI), rt = 2.82 min, m/z calcd. for C, H,,NO_,S = 521.2, found m/z = 544.2 [M+Na]* . Compound 32c was obtained as a clear oil from 31c. Yield = 81%. Compound 32d was obtained from 31d. Yield = 92%. LCMS (ESI): rt = 2.65 min, m/z calcd. for C,,H,,NO,,S = 505.1, found m/z = 528.2 [M+Na]".
Synthesis of compounds 33a, 33b, 33c, or 33d according to Formula | To a solution of 32 (a to d) (2.2 mmol, 1eq.) in MeOH (13 mL) was added NaOMe (2.2 mmol, 1.5 eq.) under an inert atmosphere of nitrogen. The reaction was stirred at room temperature under an inert atmosphere of nitrogen until completion. The reaction mixture was neutralized with Dowex (H*) and filtered. The filtrate was evaporated to dryness yielding 33a to d. Compound 33a was obtained as a brown oil from 32a. Yield = guantitative. LCMS (ESI), rt = 0.34 min, m/z calcd. for C,H, NO, S = 297.1, found m/z =
320.0 [M+Na]*. Compound 33b was obtained as a brown oil from 32b. Yield = quantitative. LCMS (ESI), rt = 0.36 min, m/z calcd. for C,H, NO.S = 311.1, found m/z =
334.0 [M+Na]* and m/z = 310.0 [M-H]. Compound 33c was obtained from 32c. Yield = 98%. Compound 33d was obtained as slightly yellow oil from 32d. Yield = quantitative. LCMS (ESI), rt = 0.34 min, m/z calcd. for C, H,,NO.S = 295.1, found m/z = 318.0 [M+Na]" and m/z = 294.0 [M-H].
The below synthesis shows the formation of specific thiol compounds 39a-c and 41 according to Formula |. First intermediate compounds 34, 35a, 35b, or 36a are prepared.
NO, | 0 0 Oy NH o 070 tg A 00; Sood, Te BON Ao > Ao °° 34 Reo N98 Aco A bas mn 363 05h
DAS / 7 OH of =X, “oT AcO re OAc AcO Yr OAc 35a 35b
OOH O fu Ae O ge ’ Sy hd = Pb U Aco” TY GAC - Ac ~~ Q et ò OAc Ah Aco“ Yr ‘Dát
DAT Lj 34 or 35a or 35b —ïgw 37a-C | I
HI 39a-c ee 383-C | | | Oy No! ON Sh ON SH AcO 0.0 AcO 0,0 HO 00 AcO “OAc AcO “OAc HO “OH OAc OAc OH 37a 38a 39a Oz NC! ON She ON SH Aco" OC Aco “00 Ho-~N-0~0 AcO “OAc AcO “OAc HO “OH OAc OAc OH 37b 38b 39b AS L x Os Nu Cl ON, She ON SH 0_,0 0. 0 ol) AcO HON Op © AcO TA ‘OAc oo) OAc Do! OH ¢ OAc OH 370 38c 39¢
/ L L Oy NH ON Cl ON -SH Aco 0 Oo Ik Aco 0 Oo Ik, HK HO 0_,0 ol) OAc ol) OAc ol To H OAc OAc OH 36a 40 41 Synthesis of intermediate compound 34 To a solution of p-nitrophenyl 2,3,4,6-tetra-O-acetyl-B-D-galactopyranosyl carbonate (3 g, 5.84 mmol, 1 eq.) in DCM (54 mL) was added a 2M solution of methylamine in THF (20.5 mL, 40.9 mmol, 7 eq.). The reaction mixture turned yellow. The solution was stirred at room temperature under an inert atmosphere of nitrogen for 20 min. The reaction mixture was diluted with DCM, washed with a 1M solution of HCI, a saturated solution of NaHCOQOs, and brine. The organic layer was dried over MgS04, filtered, and evaporated to dryness. The crude product was dissolved in DCM and purified by column chromatography (silica, 30% to 70% EtOAc in heptane) yielding 34 (2.23g, 5.5 mmol, 94%) as a white solid. LC-MS (ESI): r.t. = 2.39 min, m/z calcd. for C,H, ,NO,, =
405.1; found m/z = 428.2 [M+Na]".
Synthesis of intermediate compounds 35a-b The B-linked carbamate intermediates 35a and b were prepared from known 2,3,4,6-tetra-O-acetyl-D-galactopyranose by reaction with appropriate isocyanates (2 eq.) in toluene in the presence of triethylamine (1 eq.) for 2-24 h at 20-60°C until the starting material was completely converted into the carbamate. The reaction mixture was cooled to 15°C and 3-(dimethylamino)propylamine (1.5 eq.) was added. Stirring was continued for 30 min. The reaction mixture was extracted with 2M aq. HCI, water and aq. NaHCO3, dried on MgSO. and evaporated to give the carbamate 35a and b, which was used without further purification. Compound 35a was obtained as a white solid foam. Yield = 99%. Compound 35b was obtained as a transparent oil. Yield = 93%. Synthesis of intermediate compounds 36a-b Compounds 36a was synthesized in a similar manner as compound 35. Yield = 99%. Compounds 36b was synthesized in a similar manner as compound 35. Yield = 65%.
Synthesis of intermediate compounds 37a-c To a solution of 34 or 35a or 35b (4.44 mmol, 1 eq.) in DCM (25 mL) was added paraformaldehyde (6.65 mmol, 1.5 eq.) followed by chlorotrimethylsilane (10.6 mmol, 2.4 eq.) and the resulting solution stirred at room temperature under an inert atmosphere of nitrogen till completion. The reaction mixture was concentrated under vacuum to give 37a to c. The compounds were used as such in the next step. Compound 37a was obtained as a colorless oil. Yield = 93%. Compound 37b Yield = quantitative. Compound 37c was obtained as a white solid. Yield = quantitative. Synthesis of intermediate compounds 38a-c To a solution of 37a, b or c (8.5 mmol, 1 eq.) in DMF (20 mL) under an inert atmosphere of nitrogen was added potassium thioacetate (12.8 mmol, 1.5 eq.}. The reaction mixture was stirred under Nz at 50°C till completion. The reaction mixture was diluted with EtOAc and washed with brine (3x). It was dissolved in DCM and purified by Flash chromatography affording 38a to c. Compound 38a was obtained as an oil. Yield = quantitative. LC-MS (ESI): r.t. = 2.72 min, m/z calcd. for C,H, NO, ‚S = 493.1; found m/z = 516.2 [M+Na]*. Compound 38b was obtained as an oil. Yield = 73%. LC-MS (ESI): r.t. = 2.89 min, m/z calcd. for C,H, NO,,S = 521.2; found m/z = 544 [M+Na]*. Compound 38c was obtained as a colourless oil. Yield = 88%. LC-MS (ESI): r.t. = 2.71 min, m/z calcd. for C,H, NO, ,S = 537.2; found m/z = 560 [M+Na]*. Synthesis of compounds 39a-c according to Formula | To a solution of 38a, b or ¢ (1.1mmol, 1eq.) in MeOH (12 mL) was added NaOMe (3.3 mmol, 3eq.). The reaction was stirred at room temperature under an inert atmosphere of nitrogen till completion. The reaction mixture was then quenched with Dowex H* (pre-washed with water and MeOH), filtered and evaporated to dryness to give 39a to c. The product was used in the next step without further purification. Compound 39a was obtained as a colorless oil. Yield = quantitative. LC-MS (ESI): r.t. = 0.34 min, m/z calcd. for C,H, .NO.S = 283.1; found m/z = 306.0 [M+Na]*, m/z = 282.0 [M-H]. Compound 39b. Yield = 89%. LC-MS (ESI): r.t. = 0.34 min, m/z calcd. for C, H, NO.S =
311.1; found m/z = 334.0 [M+Na]*. Compound 39c was obtained as a colourless oil. Yield = 98%. LC-MS (ESI): r.t. = 0.54 min, m/z calcd. for C,,H,.NO,S = 327.1; found m/z =
350.0 [M+Na]*.
Synthesis of intermediate compound 40 Compounds 40 was synthesized in a similar manner as compound 37. Yield = 96%. Synthesis of compound 41 Step II-k) Compound 40 was reacted with potassium thioacetate in a similar manner as compound 37a, b or ¢ to give the corresponding thioacetate sugar intermediate. Yield = 92%. LC-MS (ESI): r.t. = 2.86 min, m/z calcd. for C,H, NOS = 521.2; found m/z = 544.2 [M+Na]*. Step III-k) To a solution of previously synthesised thioacetate sugar intermediate (100 mg,
0.19 mmol, 1eq.) in MeOH (1.9 mL) was added K:CO: (39.7 mg, 1.5 eq.) at room temperature. The reaction was stirred at room temperature under an inert atmosphere of nitrogen for 3h. The reaction mixture was then quenched with Dowex H* (pre-washed with water and MeOH), filtered and evaporated to dryness to give 41. The product was used in the next step without further purification. Compound 41 was obtained as an off- white foam. Yield = quantitative. The below synthesis shows the formation of specific thiol compound 42b. NO, o J ° oot 3 ° Oo Msn 0 90 mo osn Loko jn GAN L Ao 90 0 Lo! ° oy 070 LTA oo ok oA TT Ao °° 3° °° in 420 Synthesis of intermediate compound 42a To a solution of cystamine hydrochloride (439 mg, 1.95 mmol, 1eq.) in DCM (48 mL) was added TEA (1.63 mL, 11.7 mmol, 8 eq.) followed by the p-nitrophenyl 2,3,4,6- tetra-O-acetyl-B-D-glucopyranosyl carbonate (2 g, 3.9 mmol, 2 eq.). The solution was stirred overnight at room temperature under an inert atmosphere of nitrogen. The reaction mixture was diluted with DCM and washed with brine. The organic layer was dried, concentrated and purified by flash chromatography yielding disulfide 42a (1.49 g, 1.65 mmol, 85%) as a white solid. Synthesis of compound 42b according to Formula |
To compound 42a (150 mg, 0.17 mmol, 1 eq.) were added MeOH (0.9 mL) and DTT (81 mg, 0.51 mmol, 3 eq.). The reaction was stirred at room temperature until completion. The crude product was then concentrated and purified by column chromatography yielding compound 42b (quantitative) as a white solid. rk The below synthesis shows the formation of specific thiol compound 48 according to Formula |.
J O_ ‚OH ON Jr io 4 ln oT In Y cr Oo“ “F HO 0,0 A Cr & ol Je
OH 43 44 45 | ln / we 04 0 ~~ 0 ij OT VI-n tf T Vn, Vn Toney? Ls 2 0 5 2 0 5 or of °F 48 47 46 Synthesis of B pure anomer intermediate compound 44 2-Fluoro-2-deoxy-3,4,6-tri-O-benzyl-D-glucopyranose 43 (6g, 13.2 mmol, 1.0 eq.) (see publication European Journal of Organic Chemistry 5 (2012) 948-959), propyl isocyanate (4.69 mL, 49.6 mmol, 3.75 eq.) and TEA (3.68 mL, 26.4 mmol, 2 eq.) were dissolved in toluene (63 mL). The reaction was stirred at room temperature under an inert atmosphere of nitrogen for 45 h. 3-(Dimethylamino)-1-propylamine (4.57 mL, 36.3 mmol,
2.75 eq.) was then added to quench residual isocyanate. The reaction mixture was stirred for another 30 minutes. The mixture was diluted with EtOAc and washed with 1 M HCI, a saturated solution of NaHCO3 and brine. The organic layer was dried using MgSO.,, filtered and concentrated in vacuo. The crude product was purified by recrystalization in EtOAc/ Heptane yielding the B pure anomer 44 (6.7g, 12.5mmol, 95%) as a white crystalline solid.
LC-MS (ESI): r.t. = 3.67 min, m/z calcd. for C31H3sFNOs = 537.3, found m/z = 560.3 [M+Na]*. Synthesis of intermediate compound 45 Carbamate 44 (5.8 g, 10.9 mmol, 1.0 equiv.) was dissolved in a mixture of MeOH (46.0 mL) and EtOAc (46.0 mL) and activated carbon on Pd (410.3 mg, 3.9 mmol, 0.4 eq.) was added.
The mixture was then stirred under hydrogen atmosphere at 8 psi at room temperature for 3 days before being filtered through celite.
The solvents were evaporated under reduced pressure and the crude product was purified using column chromatography (silica, 0 to 10% MeOH in DCM) to yield 45 (2.5 g, 9.3 mmol, 85%) as white solids. °F NMR (377 MHz; MeOD): ò -201.6, -201.8; LC-MS (ESI): r.t. = 0.78 min, m/z calcd. for C19H18FNOs = 267.1, found m/z = 280 [M+Na]*. Synthesis of intermediate compound 46 Carbamate 45 (2.5 g, 9.3 mmol, 1 equiv.) was dissolved in pyridine (38.0 mL) and DMAP (113.6 mg, 0.9 mmol, 0.1 equiv.) was added.
Acetic anhydride (3.5 mL, 37.1 mmol, 4.0 equiv.) was added and the mixture was stirred for 3 hours under an inert atmosphere of nitrogen at room temperature.
After reacting, the mixture was diluted with DCM and washed with a saturated solution of NaHCO: and brine.
The organic layer was dried with MgSO, and the solvents were evaporated under reduced pressure.
The crude product was purified using column chromatography (silica, 10 to 30% EtOAc in heptane) to yield 46 (3.8 g, 9.1 mmol, 98%) as a clear oil. **F NMR (377 MHz; CDCI3): 5 -200.6 LC- MS (ESI): r.t. = 2.65 min, m/z calcd. for C16H24FNOs = 393.1, found m/z = 416.2 [M+Na]". Synthesis of intermediate compound 47 Step IV-n) To a solution of compound 46 (2.1 g, 5.34 mmol, 1eq.) in DCM (46.2 mL) was added paraformaldehyde {240 mg, 8.01 mmol, 1.5 eq.) followed by chlorotrimethylsilane (5.08 mL, 40 mmol, 7.5 eq.) and the resulting solution stirred at room temperature under an inert atmosphere of nitrogen for 6h.
The reaction mixture was then evaporated to dryness and co-evaporated with DCM.
Step V-n) The crude intermediate obtained was dissolved in DMF (21.5 mL) under Nz and potassium thioacetate (1.2 g, 10.7 mmol, 2 eq.) was added.
The reaction mixture was stirred under an inert atmosphere of nitrogen at 50°C for 1h30. The reaction mixture was then diluted with EtOAc and washed with brine, dried over MgSO4 and concentrated in vacuo.
The crude product was purified by flash chromatography (Silica, 0 to 50% EtOAc in heptane) yielding 47 (2.52 g, 5.23 mmol, 98%) as a transparent oil.
LC-MS (ESI): r.t. = 2.96 min, m/z calcd. for C1gH2sFNO1,S = 481.1, found m/z = 504.2 [M+Na]". Synthesis of compound 48 according to Formula | Sugar 47 (181.5 mg, 0.4 mmol, 1 eq.) was dissolved in MeOH (4 mL) and sodium methoxide (81.6 mg, 1.1 mmol, 3 eq.) was added.
The mixture was then stirred for 4 hours under an inert atmosphere of nitrogen at room temperature.
After completion, the reaction mixture was neutralized with Dowex H*, filtered and concentrated yielding 48 (111.3 mg, 0.36 mmol, 93%) as a transparent oil.
The product is used as such in the next step.
LC-MS (ESI): r.t. = 2.23 min, m/z calcd. for C11H20FNOsS = 313.1, found m/z = 336.0 [M+Na]*. The below schemes shows the preparation of specific thiol compounds 52a and 52b, respectively, both according to Formula I.
ON Cl ij Oy NO op Ml ON Oog AcO 00 —* AcO 00 —* AcO 0.0 AcO “OAc AcO “OAc AcO “OAc OAc OAc OAc 37b 49a 50a - N Ne ONO gy ij ON Og HON O~p © ~—— AcO 0.0 HO “OH AcO “OAc OH OAc 52a 51a
ON el ONO on ij ON OL os =X, — =X, — =X, AcO “OAc AcO “OAc AcO “OAc OAc OAc OAc 37t 48h 50b - p N 0 ON Osh OM Ons IN HO “OH AcO “OAc OH OAc 52b 51b Synthesis of intermediate compounds 49a-b To compound 37b or ¢ (11.5 mmol, 1 eq.) dissolved in dry DCM (55 mL) was added ethyleneglycol (6.4 mL, 115 mmol, 10 eq.) followed by DIPEA (10 mL, 58 mmol, 5 eq.). The reaction mixture was stirred at room temperature till completion.
The solution was diluted in EtOAc and washed with water, a 2 M solution of HCI and brine.
The organic layer was dried over MgSO4, filtered and concentrated yielding 49a and b.
Compound 49a was obtained as a white foam.
Yield = 66%. Compound 49b Yield = quantitative.
Synthesis of intermediate compounds 50a-b To compound 49a or b (1.2 g, 2.2 mmol, 1 eq.) in pyridine (15 mL) at 0 °C was slowly added MsCI (0.3 mL, 4.4 mmol, 2 eq.) The reaction was then stirred at room temperature under an inert atmosphere of nitrogen until completion.
The mixture was diluted in EtOAc, washed with water, a 2 M solution of HCI, a saturated solution of NaHCO; and brine.
The organic layer was dried over MgSO, filtered and evaporated under reduced pressure yielding 50a and b.
Compound 50a yield = quantitative.
Compound 50b yield = 97%. Synthesis of intermediate compounds 51a-b Compound 51a and b were obtained in a similar manner to that describe for compound 22. Compound 51a was obtained as an orange oil.
Yield = 92%. Compound 51b was obtained as a dark orange oil.
Yield = 60%.
Synthesis of compounds 52a-b according to Formula | Compound 52a and b were obtained in a similar manner to that describe for compound 23. Compound 52a yield = quantitative. Compound 52b yield = quantitative. The below schemes show the preparation of thiol compound 55 according to Formula I. NO, J ; / 0 5° Hg, Ig o yy" Hig, W-qg ON H Aco 00 — ed TTT Ho 00 mem Ho OH OH mem 53 54 | V-q, Vig, Vilg
J ‚ ON
HS oon
OH 55 Synthesis of intermediate compound 53 Step I-q) p-Nitrophenyl 2,3,4,6-O-acetyl-B-D-glucopyranosyl carbonate (2 mM) was dissolved in DCM (10 mL). n-Propylamine (2.5 mM) and triethylamine (2 mM) were added, and the reaction mixture was stirred overnight. The mixture was diluted with DCM and extracted successively with 1 M HCI, water and aq. NaHCO: (2 x), dried (MgSQ.) and concentrated. Purification of the residue by flash chromatography with heptane-ethyl acetate afforded the propyl carbamate (1.8 mM). Step 1l-q) The carbamate (1.8 mM) was dissolved in methanol. Sodium methoxide (0.2 mM) was added, and the mixture was stirred for 1h. Dowex H* was added and the mixture was filtered and concentrated. The resulting unprotected propyl carbamate 53 was dried in vacuo and used without further purification.
Synthesis of intermediate compound 54 Step 1lI-q) Carbamate 53 (1.8 mM) was dissolved in pyridine (10 mL), followed by the addition of TBS-CI (2.7 mM). The mixture was stirred overnight. Acetic anhydride (8 mM) was added and stirring was continued for another 18 h. Water was added, and the mixture was concentrated. The residue was diluted with ethyl acetate, extracted successively with 1M HCI, water and aq. NaHCOs, dried (MgSO.) and concentrated. The resulting oil was purified by flash chromatography with heptane-ethyl acetate to give the acetylated 6-O- TBS derivative (1.35 mM).
Step IV-q) The product from step iii (1.35 mM) was dissolved in acetonitrile (10 mL). Water (1 mL) was added, followed by solid pTsOH (4 mM). The resulting reaction mixture was stirred for 1 h. Water was added, and the mixture was extracted with DCM. The organic layer was dried (MgSO.) and concentrated. The residue was purified by flash chromatography with heptane-ethyl acetate to give the 6-hydroxy derivative 54 (1.1 mM). 1H NMR (400 MHz; CDCls): 8 3.17, dd, 1H, H-8a; 3.55, dd, 1H, H-6b; 5.67, d, 1H, H-1. Synthesis of compound 55 according to Formula | Step V-q) To a solution of 54 in toluene was added successively imidazole (3.3 mM), PPh (2.2 mM) and iodine (1.7 mM). The reaction mixture was heated at reflux for 2 h, then cooled and quenched with aq. NaHCO:. The aqueous layer was extracted with ethyl acetate and the combined organic phases were treated with I> until a brown color persisted. The organic phase was extracted with aq. Naz2S203, dried over Na2S0O.,, and concentrated under reduced pressure. The residue was purified by flash chromatography with heptane-ethyl acetate to give the 6-iodide (0.9 mM).
Step VI-q) The 6-iodide was treated with potassium thioacetate (2.7 mM) in DMF at 50° for 1h. Ethyl acetate was added, and the mixture was extracted with brine (3x). The organic layer was dried (MgSO4) and concentrated. The residue was purified by flash chromatography with heptane-ethyl acetate to give 6-thioacetate (0.75 mM).
Step VII-q) The thioacetate was dissolved in methanol. Sodium methoxide (1 mM) was added, and the reaction mixture was stirred for 1 h. Dowex H* was added and the mixture was filtered and concentrated. The residue was dried under vacuo to give 6-mercapto derivative 55 (0.75 mM). LC-MS (ESI): r.t. = 0.95 min, m/z calcd. for C1oH1sNOsS = 281.1, found m/z = 304 [M+Na]*.
Synthesis of compound 56 Compound 56 was prepared in the same way as described for 55 but using p- nitrophenyl 2,3,4,6-tetra-O-acetyl-B-D-glucopyranosyl carbonate as the starting material and diethylamine as the amine. LCMS (ESI): r.t. = 2.19 min, m/z calcd. for C+41H21NOsS =
295.1, found: m/z =318.2 [M+Na]*; m/z =294.0 [M-H]. HS oO r— © DN HO: Oo
HO OH 56 Synthesis of compound 57 Compound 57 was prepared in the same way as described for 55 but using p- nitrophenyl 2,3,4,6-tetra-O-acetyl-B-D-glucopyranosyl carbonate as the starting material and N-methyl-N-propylamine as the amine. LCMS (ESI): r.t. = 1.52 min, m/z calcd. for C11H21NQeS = 295.1; found: m/z =318.2 [M+Na]*; m/z =294.1 [M-H].
HS a + \ Sn HOw-{ 0 s
FT
HO OH 57 Synthesis of compound 58 Compound 58 was prepared in the same way, but using p-nitrophenyl 2,3,4,6- tetra-O-acetyl-B-D-glucopyranosyl carbonate as the starting material. "H NMR (400 MHz; CDs;OD): ò 5.32, d, 1H, H-1. 7 HS 0 HO $0
NS £ =
HO OH 58
SYNTHESIS OF COMPOUNDS ACCORDING TO FORMULA IVA AND IVB Disulfide Coupling of Thiosugar with alkyl-or aryl-sulfonylsulfanylmethyl-drug conjugate R1 0 sl 04 n o-visl-s— IZJDM R2a’ / R9 R1 R3 R4 Se on O—R5 Formula IVb R2a" NL RZ R4 0 Yq omMizi Hs R9 S Se N O-R5 R2a’ R3 R4 Formula Va In these prodrug compounds R1, R2a, R2b, R3, R4 and R5 are as disclosed above. In addition, for R1 (Formula IVb) all group except the thiol groups are possible, so that R1 is selected from the group consisting of -H, -F, -OH, and -O-Prot. In addition, for R5 (Formula 1Va) all groups except the thiol groups are possible, so that R5 is a C1- C6 (hetero)alkyl or -(C=O)NR7R8. Formulas IVa and IVb are specific, preferred examples of Formulas lla and IIb respectively, in which L is -S-CH(R9)-O-C(=0)-.
In Formula IVb V[S] is selected from the group consisting of -WS-, - CH2CH20CH2CH2S-, -(C=0)N(R8)YS-, -(C=0)-4-piperidine-S-, -(C=0)-3-pyrrolidine-S-, and -(C=0)-3-azetidine-S-; it should be noted that [S] is the sulfur group that is present in V: in the Formula IVb this has been denoted as such to make it clear that there is a disulfide linker between the sugar moiety and the drug moiety. V[S] is a linker group linking the sugar moiety to the drug moiety. W is a C1-C8 alkyl group, preferably -
CH2CH:- or -CH2CH2CH:-; Y is a C1-C6 (hetero)alkyl, preferably selected from the group consisting of -CHz-, -CH2CHz-, and -CH2CH2CHo»-; In Formula IVa and Formula IVb, R9 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, and t-butyl, preferably hydrogen or methyl.
DM is a drug moiety and [Z] represents a part of DM, wherein [Z] is selected from the group consisting of O, S, and N.
First Protocol — step C Synthesis of compound 59 to 61, 66, 68, 69, 72 to 80, 84 to 87 and 105 To a solution of thiosulfate-drug conjugate (1 eq.) and described thiosugar (0.38 mmol, 1 eq.) in 4 mL of a solvent, such as MeOH, THF or DMF, a solution of NaHCO; (base) (1 eq.) in water (1.8 mL) was slowly added.
The reaction mixture was stirred under an inert atmosphere of nitrogen.
After completion, the reaction mixture was then diluted with EtOAc or DCM and washed with water and brine.
The organic layer was dried, concentrated, and purified by flash chromatography to give compounds 59 to 61, 66, 68, 69, 72 to 80, 84 to 87 and 105 (according to Formula IVa and IVb). Table 2 shows the compounds that have been prepared.
Second Protocol — step c Synthesis of compound 62 to 65, 67, 70, 71, 81 to 83, 88 and 104 A solution of thiosulfate-drug conjugate (1 eq.) and described thiosugar (1.3 eq.) in a solvent, such as THF or DMF, was stirred at room temperature under an inert atmosphere of nitrogen till completion.
The reaction mixture was concentrated and purified by column chromatography yielding compounds 62 to 65, 67, 70, 71, 81 to 83, 88 and 104 (according to Formula IVa and IVb). Tables 2 show the compounds that have been prepared.
The below synthesis shows the preparation of compound 89 according to Formula IVb, in which: e R1=OH e R2a=0H e R2b=H e R3=OH e R4=F e R=H e V=(C=0)N(CsH7)CH: e [Z]=NH e DM = Gemcitabine
ON Fon m3 oF OH oet? od UID 0-si./ NCN A { nn? J oro oO NS. HON 9-9 nL HO" “F
LT OH
OH 81 89 Synthesis of compound 89 according for Formula IVb To conjugate 81 (382 mg, 0.43 mmol, 1 eq.) dissolved in MeOH (8 mL) was added a 1.25 M solution of HCI in MeOH (0.684 mL, 2eq.). The reaction mixture was stirred at room temperature for 20 min. The mixture was concentrated and precipitated from a mixture of MeOH and Et.0. The precipitate was filtered and purified by reverse phase column chromatography (RP silica, water/ACN 95/5 — 0/100) yielding the desired compound 89 (225mg, 0.34 mmol, 79%). LC-MS (ESI): r.t. = 2.39 min, m/z calcd. For C22H3:F3N4O12S2 = 6864.1; found m/z = 665.2 [M+H]*, m/z = 663.3 [M-H]. The below synthesis shows the preparation of compound 90 according to Formula IVb, in which: « R1=OH e R2a=0OH e R2b=H e R3=OH e R4=0H e V=(C=0O)NHCH2CH: e [Z]=N + DM = Cinacalcet N 5 l N A OAc Og Nas SiON _ OH Og Nag SON Cr 0 NO 0 Aco” “OAc HOY “OH OAc OH 76 90
Synthesis of compound 90 according to Formula IVb A solution of NaOMe (10 mg, 0.2 mmol, 0.5 eq.) in MeOH (5.5 mL) was added to 76 (343 mg, 0.38 mmol, 1 eq.). The resulting reaction mixture was stirred at room temperature for 20 min. The reaction mixture was neutralized with Amberlite CG 50 type 1, filtered and concentrated. The crude product was purified by flash chromatography (silica, O to 15% DCM in MeOH) to give 90 (181 mg, 0.25 mmol, 65%) as a white solid. LC-MS (ESI): r.t. = 3.20 min, m/z calcd. For C33H39F3N2OsS2 = 728.2; found m/z = 751.3 [M+Na]*, m/z = 773.2 [M-H+HCOOH].
Determination of oral bioavailability of several conjugates.
Relative and absolute bioavailability may be determined in different animal models and according to different protocols. The following protocol is typical for determining bioavailability in female Beagle dogs and was used in the present invention.
The animals were deprived from food over a time period of 8 h prior to administration of the compounds according to the present invention and over a time period of 2 h after administration of the compounds according to the present invention. Water was supplied without limitation. On the study day, the animals received the compounds according to the present invention, at a single dose of 7.5 or 15 umol/kg, by oral gavage, formulated in mixtures of propylene glycol, ethanol and 0.9% NaCl + 5% mannitol in water. Blood samples were collected from the jugular vein on the following time points: 0.25, 0.5, 1, 2, 4, 8 and 24 hours after dosing of the compounds according to the present invention. Circulating concentrations of the compounds according to the present invention were determined over a time period of 24 hours using LC-MS/MS methods with demonstrated specificity and error over a concentration range of 1.0 ng/mL (LLQ) to 2500 ng/mL (1 day validation). Pharmacokinetic parameters were calculated from concentration versus time data using non-compartmental pharmacokinetic methods using Phoenix pharmacokinetic software. Data are compared to the parent drugs to establish improvement of its oral bioavailability by the compounds according to the present invention. The following compounds have been tested:
R2b { — O-V[S]-S [Z]DM a R9 R3 R4 Table 1a shows the result. In the final column A AUC refers to the increase of AUC values for parent drugs derived from their conjugates after administration to Beagle dogs compared to AUC values obtained for the parent drugs as such after administration to Beagle dogs. This shows the effect of the use of the compounds according to the present invention. In this column: + denotes a 1.1 to 2 - fold increase of the AUC compared to parent drug; ++ denotes a 2 to 4 - fold increase of the AUC compared to the parent drug; and +++ denotes a >4 -fold increase of the AUC compared to the parent drug.
DM[Z] VS, R9 S > O—R5 R2a R3 R4 Table 1b shows the result. In the final column AAUC refers to the increase of AUC values for parent drugs derived from their conjugates after administration to Beagle dogs compared to AUC values obtained for the parent drugs as such after administration to Beagle dogs. This shows the effect of the use of the compounds according to the present invention. In this column: + denotes a 1.1 to 2 - fold increase of the AUC compared to parent drug; ++ denotes a 2 to 4 - fold increase of the AUC compared to the parent drug; and +++ denotes a >4 -fold increase of the AUC compared to the parent drug.
Table 1a: results for oral bioavailability for compounds according to the invention Com- V[S] Drug A AUC pound molecule Pe Ce a [ene | ee [nne | Re =| [Gn [oneens | A [Goa [en a|n [en [om[omems [We [ome |v [OF [ [ON |COMGRORS | [Gee |v [OR |F [Gn | CONprowIoRs | [Deme ves 83 H OH | OH | C{O)N(CH2CH2O0Me)-CH20- H | Duloxetine | +++ CT Pees Table 1b: results for oral bioavailability for compounds according to the invention Compound Drug molecule used | A J A ll =| jn comma [On [Gre |e
While specific embodiments of the subject disclosure have been discussed, the above specification is illustrative and not restrictive.
Many variations of the systems and methods herein will become apparent to those skilled in the art upon review of this specification.
The full scope of the claimed systems and methods should be determined by reference to the claims, along with their full scope of equivalents, and the specification,
along with such variations.

Claims (16)

CONCLUSIESCONCLUSIONS 1. Verbinding volgens Formule I: R1 R2b 0 \ O—R5 R2a / R3 R4 Formule | waarbij R1 is gekozen uit de groep bestaande uit -H, -F, -OH, -O-Prot, en -SH; waarbij R2a en R2b elk onafhankelijk zijn gekozen uit de groep bestaande uit -H, -OH, -O-Prot en -F; met de bepaling dat een van R2a en R2b -H is en de ander van R2a en R2b is gekozen uit de groep bestaande uit -OH, -O-Prot en -F; waarbij R3 -OH of -O-Prot is; waarbij R4 is gekozen uit de groep bestaande uit -OH, -O-Prot, en -F; waarbij R5 is gekozen uit de groep bestaande uit -WSH, -CH,CH>,OCH.CH:SH, -(CZO)N(R6)YSH, -(C=0)-4-piperidine-SH, -(C=0)-3-pyrrolidine-SH, -(C=0)-3- azetidine-SH, C1-C6 (hetero)alkyl, en -(C=O)NR7R8; waarbij Prot een beschermende groep is; waarbij R6 waterstof of een C1-C6 (hetero)alkyl is; waarbij W een C1-C6 alkylgroep is; waarbij Y een C1-C6 (hetero)alkylgroep is; waarbij R7 waterstof of een C1-C6 alkyl is; waarbij R8 een C1-C6 alkylgroep is; met de bepaling dat indien R1 -SH is, R5 dan een C1-C86 (hetero)alkyl of -(C=O)NR7R8 is; met de verdere bepaling dat indien R5 is gekozen uit de groep bestaande uit -YSH, -CH2CH:OCH:CH2SH, -{C=O)N(R6)WSH, -(C=0)-4-piperidine- SH, -(C=0)-3-pyrrolidine-SH, en -C(=0)-3-azetidine-SH, dan is R1 gekozen uit de groep bestaande uit of -H, -F, -OH, en -O-Prot.1. Compound of Formula I: R1 R2b 0 \ O—R5 R2a / R3 R4 Formula | wherein R1 is selected from the group consisting of -H, -F, -OH, -O-Prot, and -SH; wherein R2a and R2b are each independently selected from the group consisting of -H, -OH, -O-Prot and -F; with the provision that one of R2a and R2b is -H and the other of R2a and R2b is selected from the group consisting of -OH, -O-Prot and -F; wherein R3 is -OH or -O-Prot; wherein R4 is selected from the group consisting of -OH, -O-Prot, and -F; wherein R5 is selected from the group consisting of -WSH, -CH,CH>,OCH.CH:SH, -(CZO)N(R6)YSH, -(C=O)-4-piperidine-SH, -(C =O)-3-pyrrolidine-SH, -(C=O)-3-azetidine-SH, C1-C6 (hetero)alkyl, and -(C=O)NR7R8; wherein Prot is a protecting group; wherein R6 is hydrogen or a C1 -C6 (hetero)alkyl; wherein W is a C1-C6 alkyl group; wherein Y is a C1-C6 (hetero)alkyl group; wherein R7 is hydrogen or a C1-C6 alkyl; wherein R8 is a C1-C6 alkyl group; with the proviso that if R1 is -SH then R5 is a C1-C86 (hetero)alkyl or -(C=O)NR7R8; with the further provision that if R5 is selected from the group consisting of -YSH, -CH2CH:OCH:CH2SH, -{C=O)N(R6)WSH, -(C=O)-4-piperidine-SH, - (C=O)-3-pyrrolidine-SH, and -C(=O)-3-azetidine-SH, then R1 is selected from the group consisting of or -H, -F, -OH, and -O-Prot . 2. Verbinding volgens conclusie 1, waarbij elke Prot een beschermende groep is onafhankelijk gekozen uit de groep bestaande uit acetyl, benzoyl, benzyl, 4-methoxybenzyl, trialkylsilyl, bij grootste voorkeur acetyl.A compound according to claim 1, wherein each Prot is a protecting group independently selected from the group consisting of acetyl, benzoyl, benzyl, 4-methoxybenzyl, trialkylsilyl, most preferably acetyl. 3. Verbinding volgens een van de voorgaande conclusie, waarbij R5 -(C=0)N(R6)YSH is en waarbij RS is gekozen uit de groep bestaande uit waterstof, methyl, ethyl, n-propyl en -CH:CHz2OCHa.A compound according to any preceding claim wherein R5 is -(C=O)N(R6)YSH and wherein R5 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl and -CH:CH22OCha. 4. Verbinding volgens een van conclusies 1 tot 3, waarbij R5 -WSH is en waarbij W -CH:sCH- of -CH2CH2CH-- is.A compound according to any one of claims 1 to 3, wherein R 5 is -WSH and wherein W is -CH: 5 CH- or -CH 2 CH 2 CH--. 5. Verbinding volgens een van conclusies 1 tot 3, waarbij R5 -(C=O)N(R6)YSH is en waarbij Y is gekozen uit de groep bestaande uit -CH2-, -CH2CHz-, en -CH2CH2CHz-.A compound according to any one of claims 1 to 3, wherein R 5 is -(C=O)N(R 6 )YSH and wherein Y is selected from the group consisting of -CH 2 -, -CH 2 CH 2 -, and -CH 2 CH 2 CH 2 -. 6. Verbinding volgens een van conclusies 1 tot 3, waarbij R5 -(C=O)NR7R8 is en waarbij R7 is gekozen uit de groep bestaande uit waterstof, methyl, ethyl, en n-propyl.A compound according to any one of claims 1 to 3 wherein R5 is -(C=O)NR7R8 and wherein R7 is selected from the group consisting of hydrogen, methyl, ethyl, and n-propyl. 7. Verbinding volgens een van de voorgaande claim, waarbij R5 -(C=O)NR7R8 is en waarbij R8 is gekozen uit de groep bestaande uit methyl, ethyl, en n-propyl.A compound according to any preceding claim wherein R5 is -(C=O)NR7R8 and wherein R8 is selected from the group consisting of methyl, ethyl, and n-propyl. 8. Verbinding volgens een van de voorgaande conclusies, waarbij ten minste een van R1, R2a, R2b of R4 -F is.A compound according to any one of the preceding claims, wherein at least one of R1, R2a, R2b or R4 is -F. 9. Verbinding volgens conclusie 4, waarbij indien W -CHsCHs- is onder voorwaarde dat: R1 -H of -F is; en/of R2a en R2b elk onafhankelijk -H of -F zijn, onder voorwaarde dat een van R2a en R2b -H is en de ander van R2a en R2b -F is; en/of R4 -F is.The compound of claim 4 wherein when W is -CH 3 CH 3 - provided that: R 1 is -H or -F; and/or R2a and R2b are each independently -H or -F, provided that one of R2a and R2b is -H and the other of R2a and R2b is -F; and/or R4 is -F. 10. Verbinding van conclusie 1, gekozen uit de groep bestaande uit:The compound of claim 1 selected from the group consisting of: 8H o N 0 \ SH ; o_ ‚0 Hor” HO 9% 0 HO Fy OH HO “OH Ho “OH OH dn OH SH SH | o AT o AT Og NSH oO o 0 O 0 O “LX HO HO 7 " I," HO "OH HOT OH8H o N 0 \ SH ; o_ ‚0 Hor” HO 9% 0 HO Fy OH HO “OH Ho “OH OH dn OH SH SH | o AT o AT Og NSH oO o 0 O 0 O “LX HO HO 7 " I," HO "OH HOT OH OH OH N T, J Oy NSH ON SH OM. SH HO 9 0 0_.0 00OH OH N T, J Oy NSH ON SHOM. SH HO 9 0 0_.0 00 LT HO Y HO HO “OH HO" "OH HOY NFLT HO Y HO HO “OH HO" "OH HOY NF OH OH OH | | HS o ~ J °o0 JN ON Orsi O NH HOw Oo o_ 0 { HO y ) | HE" o_ 0 HO OH HO “OH u OH HO "OHOH OH OH | | HS o ~ J °o0 JN ON Orsi O NH HOw Oo o_ 0 { HO y ) | HE" o_ 0 HO OH HO “OH u OH HO "OH OH £ ~OH£~ D H o Á H Yam A o ON rg 9 ) N ON 0 __~ A 0.0 Hud Som x ~~ TSH © OJ L Jl HON OG 007 0 HO OH HO “OH 07 Ou” OH 0D H o Á H Yam A o ON rg 9 ) N ON 0 __~ A 0.0 Hud Sum x ~~ TSH © OJ L Jl HON OG 007 0 HO OH HO “OH 07 Ou” OH 0 ON SH HO 0_,0ON SH HO 0_.0 HOY OHHOY OH OHOH 11. Werkwijze voor het bereiden van een verbinding volgens Formule | volgens conclusie 1, de werkwijze omvattende:11. Process for preparing a compound of Formula | according to claim 1, the method comprising: RT’RT' Se fi pe «(}—R§' ———— s Formule!Se fi pe «(}—R§' ———— s Formula! R2a" \ R3 R4R2a" \ R3 R4 Formule Ib waarbij: R1' is gekozen uit de groep bestaande uit -H, -F, -OH, -O-Prot, en X; R2a, R2b, R3 en R4 zijn zoals bepaald in conclusie 1;Formula Ib where: R1' is selected from the group consisting of -H, -F, -OH, -O-Prot, and X; R2a, R2b, R3 and R4 are as defined in claim 1; R5' is gekozen uit de groep bestaande uit -W-X, -CH2CHzOCH:CH:X, - (C=O)N(R6)YX, -(C=0)-4-piperidine-X, -(C=0)-3-pyrrolidine-X, -(C=0)-3-azetidine-X, C1-C6 (hetero)alkyl, en -C{=O)NR7R8;R5' is selected from the group consisting of -W-X, -CH2CH2OCH:CH:X, -(C=O)N(R6)YX, -(C=O)-4-piperidine-X, -(C=0) -3-pyrrolidine-X, -(C=O)-3-azetidine-X, C1-C6 (hetero)alkyl, and -C{=O)NR7R8; X is methaansulfonaat of een halide gekozen uit de groep bestaande uit chloor,X is methane sulfonate or a halide selected from the group consisting of chlorine, broom, en jood;bromine, and iodine; R6 waterstof of een C1-C6 (hetero)alkyl is;R6 is hydrogen or a C1 -C6 (hetero)alkyl; W een C1-C8 alkylgroep is; W is een linkergroep dat de suikerring aan een halide of methaansulfonaatgroep koppelt;W is a C1-C8 alkyl group; W is a linker group linking the sugar ring to a halide or methanesulfonate group; Y een C1-C6 (hetero)alkyl is; Y is een linkergroep die de amidegroepY is a C1 -C6 (hetero)alkyl; Y is a linker group connecting the amide group ((C=O)N(RS8)) aan de halidegroep koppelt;((C=O)N(RS8)) to the halide group; R7 waterstof of een C1-C6 alkyl is;R7 is hydrogen or a C1 -C6 alkyl; R8 een C1-C6 alkyl is; onder voorwaarde dat;R8 is a C1 -C6 alkyl; provided that; in geval R1' X is, dan is R5' C1-C6 (hetero)alkyl of -(C=0Q)NR7RS;in case R1' is X then R5' is C1-C6 (hetero)alkyl or -(C=OQ)NR7RS; in geval R5' is gekozen uit de groep bestaande uit -W-X, -CH,CH>OCH,>CH:X, -(C=0)N(RB)YX, -(C=0)-4-piperidine-X, -(C=0)-3-pyrrolidine-X, en -{C=0)-3- azetidine-X, dan is R1' gekozen uit de groep bestaande uit -H, -F, -OH, en -O-Prot;when R5' is selected from the group consisting of -W-X, -CH,CH>OCH,>CH:X, -(C=O)N(RB)YX, -(C=O)-4-piperidine-X , -(C=O)-3-pyrrolidine-X, and -{C=O)-3-azetidine-X, then R1' is selected from the group consisting of -H, -F, -OH, and -O -prot; waarbij de werkwijze de volgende stappen i) en ii) omvat:the method comprising the following steps i) and ii): stap i) omvat het reageren van een verbinding van Formule Ib met kaliumthioacetaat om een thioacetaat-adduct van de verbinding Formule Ib te verkrijgen; en stap ii) omvat het reageren van het thioacetaat-adduct van de verbinding Formule Ib verkregen in stap i) met natriummethoxide, kaliumcarbonaat of natriummethaanthiolaat om de verbinding van Formule | te verkrijgen.step i) comprises reacting a compound of Formula Ib with potassium thioacetate to obtain a thioacetate adduct of the compound of Formula Ib; and step ii) comprises reacting the thioacetate adduct of the compound of Formula Ib obtained in step i) with sodium methoxide, potassium carbonate or sodium methanethiolate to give the compound of Formula | to obtain. 12. Werkwijze volgens conclusie 11, verder omvattende een stap 0), voorafgaand aan stap i), de stap 0) omvattende het reageren van een verbinding volgens Formule [1 RT’ R2b 0 ox _(Q)-Prot R2a" \ _/ R3 R4 Formule II! met een broomalcohol met formule Br-(C1-C8)-alkyl-OH, om een verbinding volgens Formule Ib te verkrijgen waarbij R5' -W-X, waarin W (C1-C6)-alkyl is, en waarin X Br is.The method of claim 11, further comprising a step 0), prior to step i), step 0) comprising reacting a compound of Formula [1 RT' R2b 0 ox _(Q)-Prot R2a" \ _/ R3 R4 Formula II with a bromoalcohol of formula Br-(C1-C8)-alkyl-OH, to obtain a compound of Formula Ib wherein R5' -W-X, where W is (C1-C6)-alkyl, and where X Br is. 13. Gebruik van een verbinding volgens een van conclusies 1-10 of verkregen door de werkwijze van conclusie 11 of 12 als een promoiety bevestigd aan een geneesmiddelmolecuul om een prodrug te verkrijgen, om de orale biologische beschikbaarheid van het geneesmiddelmolecuul te verbeteren.Use of a compound according to any one of claims 1-10 or obtained by the method of claim 11 or 12 as a promoiety attached to a drug molecule to obtain a prodrug, to improve the oral bioavailability of the drug molecule. 14. Gebruik van de verbinding volgens een van conclusies 1-10 of verkregen door de werkwijze van conclusie 11 of 12 voor de bereiding van een verbinding volgens Formule Ila of een farmaceutisch aanvaardbaar zout daarvan: DM[Z]—L—S R2b 0 D O—R5 R2a / R3 R4 Formule lla of volgens Formule IIb of een farmaceutisch aanvaardbaar zout daarvan:Use of the compound of any one of claims 1-10 or obtained by the process of claim 11 or 12 for the preparation of a compound of Formula IIa or a pharmaceutically acceptable salt thereof: DM[Z]-L-S R2b 0 D O —R5 R2a / R3 R4 Formula 11a or according to Formula IIb or a pharmaceutically acceptable salt thereof: R1 R2b O > O—V—L—[Z]DM R2a / R3 R4 Formule IIb waarbij R1 is gekozen uit de groep bestaande uit -H, -F, -OH, en -O-Prot; waarbij R5 een C1-C86 (heterojalkyl of -{C=O)NR7R8 is; waarbij V is gekozen uit de groep bestaande uit -WS-, -CH2CH2OCH2:CH:S-, - (CZO)N(R6)YS-, -(C=0)-4-piperidine-S-, -(C=0)-3-pyrrolidine-S-, en -(C=0)-3- azetidine-S-; waarbij R2a, R2b, R3, R4, R6, R7, R8, W, en Y zijn geopenbaard in conclusie 1; waarbij L een linkermolecuul is, bij voorkeur -S-CH(R8)-O-C(=0); waarbij R9 is gekozen uit de groep bestaande uit waterstof, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, en t-butyl, bij voorkeur waterstof of methyl; waarbij DM een drug moiety is en [Z] een deel van DM vertegenwoordigt; en waarbij Z is gekozen uit de groep bestaande uit O, S, en N.R1 R2b O > O—V—L—[Z]DM R2a / R3 R4 Formula IIb where R1 is selected from the group consisting of -H, -F, -OH, and -O-Prot; wherein R5 is C1-C86(heteroylalkyl or -{C=O)NR7R8; where V is selected from the group consisting of -WS-, -CH2CH2OCH2:CH:S-, -(CZO)N(R6)YS-, -(C=O)-4-piperidine-S-, -(C= O)-3-pyrrolidine-S-, and -(C=O)-3-azetidine-S-; wherein R2a, R2b, R3, R4, R6, R7, R8, W, and Y are disclosed in claim 1; wherein L is a linker molecule, preferably -S-CH(R8)-O-C(=O); wherein R9 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, and t-butyl, preferably hydrogen or methyl; where DM is a drug moiety and [Z] represents part of DM; and wherein Z is selected from the group consisting of O, S, and N. 15. Verbinding volgens Formule lla of Formule Ilb of een farmaceutisch aanvaardbaar zout daarvan R1 PHATE o R2b oO \ O_RS D O—V—L—[Z]DM R22" \_ R2a / ri Ra R3 R4 Formule lla Formula IIb waarbij R1 is gekozen uit de groep bestaande uit -H, -F, -OH, en -O-Prot; waarbij R5 een C1-C6 (hetero)alkyl of -(C=O)NR7R8 is; waarbij V is gekozen uit de groep bestaande uit -WS-, -CH2CH2OCH2CH2S-, - (CZO)N(RS8)YS-, -(C=0)-4-piperidine-S-, -(C=0)-3-pyrrolidine-S-, en -{C=0)-3- azetidine-S-;15. Compound of Formula Ila or Formula Ilb or a pharmaceutically acceptable salt thereof R1 PHATE o R2b oO \ O_RS D O—V—L—[Z]DM R22" \_ R2a / ri Ra R3 R4 Formula Ila Formula IIb where R1 is selected from the group consisting of -H, -F, -OH, and -O-Prot, where R5 is a C1-C6 (hetero)alkyl or -(C=O)NR7R8, where V is selected from the group consisting of - WS-, -CH2CH2OCH2CH2S-, -(CZO)N(RS8)YS-, -(C=O)-4-piperidine-S-, -(C=O)-3-pyrrolidine-S-, and -{C =O)-3-azetidine-S-; waarbij R2a, R2b, R3, R4, R6, R7, R8, W, en Y zijn geopenbaard in conclusie 1; waarbij L een linkermolecuul is, bij voorkeur -S-(C1-C6 alkyl)-O-C(=0)- of -S- CH(R9)-O-C(=0); waarbij R9 is gekozen uit de groep bestaande uit waterstof, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, en t-butyl, bij voorkeur waterstof of methyl; waarbij DM een drug moiety is en [Z] een deel van DM vertegenwoordigt; en waarbij Z is gekozen uit de groep bestaande uit O, S, en N.wherein R2a, R2b, R3, R4, R6, R7, R8, W, and Y are disclosed in claim 1; wherein L is a linker molecule, preferably -S-(C1 -C6 alkyl)-O-C(=O)- or -S-CH(R9 )-O-C(=O); wherein R9 is selected from the group consisting of hydrogen, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, and t-butyl, preferably hydrogen or methyl; where DM is a drug moiety and [Z] represents part of DM; and wherein Z is selected from the group consisting of O, S, and N. 16. Verbinding volgens conclusie 15 voor gebruik als een medicijn, therapie, beeldvormend middel of diagnostisch middel.A compound according to claim 15 for use as a medicine, therapy, imaging agent or diagnostic agent.
NL2028505A 2021-06-21 2021-06-21 Sugar thiol compounds and methods of producing the same NL2028505B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NL2028505A NL2028505B1 (en) 2021-06-21 2021-06-21 Sugar thiol compounds and methods of producing the same

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
NL2028505A NL2028505B1 (en) 2021-06-21 2021-06-21 Sugar thiol compounds and methods of producing the same

Publications (1)

Publication Number Publication Date
NL2028505B1 true NL2028505B1 (en) 2022-12-29

Family

ID=77911056

Family Applications (1)

Application Number Title Priority Date Filing Date
NL2028505A NL2028505B1 (en) 2021-06-21 2021-06-21 Sugar thiol compounds and methods of producing the same

Country Status (1)

Country Link
NL (1) NL2028505B1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000064485A2 (en) * 1999-04-28 2000-11-02 Genencor International, Inc. Specifically targeted catalytic antagonists and uses thereof
JP2001231593A (en) * 2000-02-25 2001-08-28 Osaka City Novel glycoside and a method of producing the same
US20050130240A1 (en) * 2003-02-19 2005-06-16 Academia Sinica Carbohydrate encapsulated nanoparticles
WO2010077623A1 (en) 2008-12-08 2010-07-08 Sanofi-Aventis Intermediates and processes for the preparation of fluoroglycoside derivatives

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000064485A2 (en) * 1999-04-28 2000-11-02 Genencor International, Inc. Specifically targeted catalytic antagonists and uses thereof
JP2001231593A (en) * 2000-02-25 2001-08-28 Osaka City Novel glycoside and a method of producing the same
US20050130240A1 (en) * 2003-02-19 2005-06-16 Academia Sinica Carbohydrate encapsulated nanoparticles
WO2010077623A1 (en) 2008-12-08 2010-07-08 Sanofi-Aventis Intermediates and processes for the preparation of fluoroglycoside derivatives

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
"Prodrugs and Targeted Delivery", 2011, WILEY-VCH
ALTERN. LAB. ANIM., vol. 29, 2001, pages 649 - 668
APFEL ULF-PETER ET AL: "Functionalized sugars as ligands towards water-soluble [Fe-only]hydrogenase models", EUROPEAN JOURNAL OF INORGANIC CHEMISTRY, WILEY-VCH VERLAG , WENHEIM, DE, vol. 2008 (32), no. 32, 1 January 2008 (2008-01-01), pages 5112 - 5118, XP009533687, ISSN: 1434-1948, Retrieved from the Internet <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/ejic.200800720> [retrieved on 20081103], DOI: 10.1002/EJIC.200800720 *
ASHISH A KULKARNI ET AL: "Glycan Encapsulated Gold Nanoparticles Selectively Inhibit Shiga Toxins 1 and 2", BIOCONJUGATE CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 21, no. 8, 18 August 2010 (2010-08-18), pages 1486 - 1493, XP002696214, ISSN: 1043-1802, [retrieved on 20100729], DOI: 10.1021/BC100095W *
BIOORG. MED. CHEM. LETT., vol. 26, 2016, pages 3774
EUROPEAN JOURNAL OF ORGANIC CHEMISTRY, vol. 5, 2012, pages 948 - 959
GARCIA-BARRIENTOS A ET AL: "Synthesis of Beta-Cyclodextrin, Per-O-glycosylated through an Ethylene Glycol Spacer Arm", SYNTHESIS, GEORG THIEME VERLAG, STUTTGART, DE, vol. 7, 1 January 2001 (2001-01-01), pages 1057 - 1064, XP002980810, ISSN: 0039-7881, DOI: 10.1055/S-2001-14574 *
JOHARY N S ET AL: "Dithiols. XV. Some polyhydroxy derivatives of 3,4- dimercaptobutanol", JOURNAL OF THE CHEMICAL SOCIETY,, 1 January 1955 (1955-01-01), pages 1292 - 1298, XP008144720, ISSN: 0368-1769 *
LOAIZA OSCAR A ET AL: "Nanostructured Disposable Impedimetric Sensors as Tools for Specific Biomolecular Interactions: Sensitive Recognition of Concanavalin A", ANALYTICAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 83, no. 8, 15 April 2011 (2011-04-15), pages 2987 - 2995, XP009533689, ISSN: 0003-2700, DOI: 10.1021/AC103108M *
TAKAHASHI T ET AL: "Synthesis of 9-Membered Masked Enediyne Analogues Possessing DNA Intercalator and Sugar Moieties", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, ELSEVIER, AMSTERDAM, NL, vol. 8, no. 23, 1 December 1998 (1998-12-01), pages 3299 - 3302, XP004143746, ISSN: 0960-894X, DOI: 10.1016/S0960-894X(98)00605-2 *
VALEPYN EMMANUEL ET AL: "Lipase-catalyzed synthesis of L-cysteine glucosyl esters in organic mediaValepyn", BIOCATALYSIS AND BIOTRANSFORMATION,, vol. 29, no. 1, 1 January 2011 (2011-01-01), pages 25 - 30, XP009533688, ISSN: 1029-2446, DOI: 10.3109/10242422.2010.548555 *
VON BURG R ET AL: "Biliary mobilization of cadmium by 2,3-dimercaptopropanol and some related compounds", JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH, TAYLOR 6 FRANCIS, US, vol. 6, no. 1, 1 January 1980 (1980-01-01), pages 75 - 85, XP009533681, ISSN: 0098-4108, DOI: 10.1080/15287398009529832 *

Similar Documents

Publication Publication Date Title
EP3250581B1 (en) Alpha-d-galactoside inhibitors of galectins
US20220185838A1 (en) Carbohydrate ligands that bind to igm antibodies against myelin-associated glycoprotein
JPS61148193A (en) Tumor treatment agent
US11414451B2 (en) Floxuridine synthesis
EP3419959A1 (en) Combination therapy
US11633484B2 (en) Method for improving the oral bioavailability of a drug
EP3404033B1 (en) C-glucoside derivative containing fused phenyl ring or pharmaceutically acceptable salt thereof, process for preparing same, and pharmaceutical composition comprising same
Manta et al. Synthesis of 4, 6-dideoxy-3-fluoro-2-keto-β-D-glucopyranosyl analogues of 5-fluorouracil, N6-benzoyl adenine, uracil, thymine, N4-benzoyl cytosine and evaluation of their antitumor activities
NL2028505B1 (en) Sugar thiol compounds and methods of producing the same
EP3256482B1 (en) Process for the preparation of sglt inhibitor compounds
WO2002057216A1 (en) Terphenyl compounds bearing substituted amino groups
US11667661B2 (en) Benzimidazole derivatives, preparation method thereof and use thereof as anti-cancer agent comprising the same
US10703772B2 (en) Processes for the preparation of SGLT-2 inhibitors, intermediates thereof
US20100081708A1 (en) Anticoagulant compounds
US11548906B2 (en) Processes for the preparation of SGLT-2 inhibitors, intermediates thereof
NL2028506B1 (en) Prodrugs with 1-(disulfanyl)alkyloxy-carbonyl units
WO2023072292A1 (en) Efficient antiviral compound and use thereof
CN113171467B (en) Chimeric molecule based on NQO1 regulation and control and application thereof
US8168766B2 (en) Process of making 2-deoxy-2,2-difluoro-D-ribofuranosyl nucleosides and intermediates therefor
CN114599643B (en) Aryl glucoside derivative
US10925887B2 (en) Steroid saponins with anti-cancer activity
KR101241321B1 (en) Improved preparation method of Decitabine
WO2017101785A1 (en) Compound, preparation method therefor, pharmaceutical composition thereof and use thereof
EP3381910B1 (en) 4-oxo-4,5-dihydrothiazole derivative, preparation method and use thereof
US20180296507A1 (en) Redox-activated glycoconjugated prochelators