Method for treating acute myeloid leukemia

文档序号:1357752 发布日期:2020-07-24 浏览:30次 中文

阅读说明:本技术 治疗急性髓系白血病的方法 (Method for treating acute myeloid leukemia ) 是由 蒂娜·拉芙兰诺斯 于 2018-10-17 设计创作,主要内容包括:发明涉及一种利用对其有用的药物治疗急性髓性白血病(AML)的方法。所述药物可以是药物组合物或套装,其包含当前描述的式(I)的化合物或其盐、溶剂化物或前药。本发明的特定化合物包括2-甲基-7-羟基-3-(3,4,5-三甲氧基苯甲酰基)-6-甲氧基苯并呋喃,也称为BNC105,以及6-甲氧基-2-甲基-3-(3,4,5-三甲氧基苯甲酰基)苯并呋喃-7-磷酸二氢钠,也称为BNC105P。(The invention relates to a method of treating acute myeloid leukemia (AM L) with a drug useful therefor, which can be a pharmaceutical composition or kit comprising a compound of formula (I) as presently described or a salt, solvate or prodrug thereof specific compounds of the invention include 2-methyl-7-hydroxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran, also known as BNC105, and 6-methoxy-2-methyl-3- (3,4, 5-trimethoxybenzoyl) benzofuran-7-dihydrogenphosphate, also known as BNC 105P.)

1.A method for treating acute myeloid leukemia (AM L) in a patient, comprising the step of administering an effective amount of a compound of formula (I):

2. the method of claim 1, wherein the patient is a human subject.

3. The method of claim 1, wherein the effective amount administered is about 0.05 to 18mg/kg body weight/day.

4. The method of claim 1, wherein the effective amount administered is from about 0.4mg to 16mg/kg body weight/day.

5. The method of claim 1, wherein the compound is administered by parenteral administration.

6. The method of claim 1, wherein the compound is co-administered with another anti-neoplastic agent, either simultaneously or sequentially.

7. The method of claim 1, wherein the patient is selected for treatment based on clinical parameters including age, level of progression of the disease, and/or other complications.

8. Use of a compound of formula (I) or a salt, solvate or prodrug thereof in the manufacture of a medicament for treating AM L in a patient, the formula (I) being:

9. a compound of formula (I) or a salt, solvate or prodrug thereof, for use in treating AM L in a patient, the formula (I) being:

10. the use of claim 8 or the compound of claim 9, wherein the patient is a human.

11. The use as claimed in claim 8 or a compound as claimed in claim 9, wherein the compound is used in combination with another anti-tumour agent.

12. A pharmaceutical kit for treating AM L comprising a compound of formula (I) and instructions for use, the formula (I) being as follows:

13. a method, use or kit according to any one of claims 1 to 12 wherein the compound of formula (I) is:

Disclosure of Invention

The present disclosure enables effective treatment of AM L, based in part on the use of a compound of formula (I) or a salt, solvate or prodrug thereof, in selectively and preferentially inducing apoptosis of AM L the compound of formula (I) is as follows:

the compound of formula (I) [ 2-methyl-7-hydroxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran ] can be prepared by the synthetic method described in PCT/AU2007/000101(WO 07/087684), the contents of which are incorporated by reference.

Accordingly, provided herein is a method for treating acute myeloid leukemia (AM L) in a patient, comprising the step of administering an effective amount of a compound of formula (I):

also taught herein is the use of a compound of formula (I), or a salt, solvate, or prodrug thereof, in the manufacture of a medicament for treating a patient suffering from acute myeloid leukemia (AM L), said formula (I) being as follows:

in a related embodiment, the description has instructive significance to a compound of formula (I), or a salt, solvate, or prodrug thereof, for use in treating AM L in a patient, said formula (I) being as follows:

in another embodiment, the method comprises treating a subject in need thereof with an effective amount of a compound of formula (I) to induce apoptosis in AM L cells.

Reference to "AM L" includes its subtypes and related forms.

Brief description of the drawings

FIG. 1 relative fluorescence measured by CellTox Green assay cytotoxicity (A) and by CellTiter-Blue assay viability (B) in the indicated dose of EX1 treated AM L cell line 48h IC50 dose was determined by the absolute IC50 method in Prism 6N 2 for MV 4; 11 and MO L M13 in both assays and n 1 for the remaining cell lines in both assays.

FIG. 2A. Total ROS levels measured by CM-H2DCFDA fluorescence after EX1 treatment of OCI-AM L324 hours 324 values represent Mean Fluorescence Intensity (MFI) relative to untreated (DMSO) control.

B. Mitochondrial O2-level, measured by MitoSox fluorescence, 24 hours after EX1 treatment of the cell line. Values represent Mean Fluorescence Intensity (MFI) relative to untreated (DMSO) control.

FIG. 3 cell cycle distribution of cell lines after 24 hours of treatment with the indicated dose of EX1 MV4, 11 and MO L M13 showed significant apoptosis induction (sub-G0 gate) and a decrease in G2/M under the action of 10nM drug KG1, H L60, ME1 and OCI-AM L3 showed G2/M cell cycle arrest KG1 and H L60 also showed an increase in the sub-G0 population starting from treatment with 10nM EX 1.

FIG. 4: western blots were performed with indicated doses of EX1 to treat protein lysates of 8h and 24h cell lines. Induction of P-JNK KT in all cell lines with 10nM EX1 at 8h183/Y185Noxa induction was observed only in 8h OCI-AM L3 and ME1 lines treated with 10nM EX1 HEK293(HEK) protein lysate was used as a positive control for Noxa detection tubulin was used as a loading control.

Figure 5 Caspase 3/7 activity (doses listed in legend in nM) was measured in raw MNC samples from Healthy Control (HC) and AM L samples after treatment with EX1 or OXi4503 after treatment with EX1 or OXi4503 for 48h (c) and 72h (d) and after staining with Annexin V/7AAD on the same raw samples viability was assessed by flow cytometry.

FIG. 6A. representative dot plots for flow cytometry analysis for detecting the population (Green Gate) comprising GPR56+ L2 SC in each of the AM L samples used (AM L136, AM L0235 and AM L1480). B. the percentage of GPR56+ cells determined by the Green gate on A of the three AM L3 samples 48 hours after treatment with EX1 or OXi 4503. C. the relative amount of L5 SC determined on AM L4 samples by immunophenotypic flow cytometry analysis 48 hours after treatment with EX1 or OXi 4503. L SC gating strategy AM L136 and AM L480 are GPR56+/CD34+ and GPR56+/CD34-, AM L235 is GMPR56+/CD34+, AM L086 and AM L138 are AM L372, CD 34/CD 38-/CD93+ is SC L.

FIG. 7 colony formation was blocked after treatment of L-AM L6 (A) and L-AM L419 (B) with the indicated doses of EX1 OXi4503 reduced colony formation on L-AM L6 at 100nM, but completely inhibited colonies both at the highest dose of this sample and at both doses L-AM L419.

FIG. 8 Western blot 48h immunoblots of L-AM L6 protein lysates from AM L samples treated with indicated doses of EX1 or OXi 4503P-JNK KTT183/Y185Induced by 500nM EX1, but not by OXi 4503. The expression of Noxa is very low and does not appear to change with any treatment. HEK293(HEK) protein lysate was used as a positive control for the Noxa assay.

Tubulin was used as loading control.

FIG. 9 MTS proliferation assay performed showed that EX1 was at least 10-fold more potent in H L-60 and MV4-11 cell lines than any other VDA after 72H exposure to drug.

DETAILED DESCRIPTIONS

Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as, an acknowledgment or admission or any form of suggestion that prior publication (or information) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.

As used in this specification, the singular forms "a," "an," and "the" include plural aspects unless the context clearly dictates otherwise.

The present disclosure teaches that compounds of formula (I) are potent Tubulin Polymerization Inhibitors (TPI) that induce tumor cell death by up-regulating pro-apoptotic proteins. An important aspect of the compounds of formula (I) is that the combination of specific C-6 and C-7 substituents with a C-2Q-group (especially C-2 methyl) appears to have higher potency and selectivity when compared to other structurally related TPI compounds.

It will be appreciated that the compound of formula (I) may be administered to a subject in the form of a pharmaceutically acceptable salt thereof. Suitable pharmaceutically acceptable salts include, but are not limited to, salts of pharmaceutically acceptable inorganic acids, such as hydrochloric, sulfuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids, or salts of pharmaceutically acceptable organic acids, such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, maleic, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulfonic, toluenesulfonic, benzenesulfonic, sulfosalicylic, aspartic, glutamic, ethylenediaminetetraacetic, stearic, palmitic, oleic, lauric, pantothenic, tannic, ascorbic and valeric acids.

Base salts include, but are not limited to, salts with pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, ammonium, and alkylamines. In one embodiment, the methods described herein include within their scope cationic salts, such as sodium or potassium salts or alkyl esters (e.g., methyl, ethyl) of phosphate.

It is also understood that any compound that is a prodrug of the compound of formula (I) is also within the scope and spirit of the treatment regimens described herein. The term "prodrug" is used in its broadest sense and includes those derivatives that are converted in vivo to the compounds of the invention (e.g., compounds of formula (I)). Such derivatives will readily occur to those skilled in the art and include, for example, those in which the hydroxyl group is free (e.g., at the C-7 position or R)1D) Compounds converted into esters, e.g. acetates or phosphates, or in which the free amino group (e.g. at C-7 or R)1D) The steps of being converted to an amide (e.g., α -amino acid amide) for esterification, e.g., acylation, of a compound are well known in The art and may include treating The compound with a suitable carboxylic acid, anhydride, or chloride in The presence of a suitable catalyst or base one prodrug is a disodium phosphate ester of The compound of The invention (e.g., The disodium C-7 phosphate ester of The compound of formula (I)) in increasing The solubility of The compound, e.g., this would allow delivery of The compound in a benign vehicle such as saline. The disodium phosphate ester may be prepared according to The methods described in Pettit, G.R., et al, Anticancer Drug drugs Des (antineoplastic Design), 1995,10,299 other documents generally describing prodrugs (and preparation thereof) including Design of prodrugs, 1985, H.Bundggaard (Eldgprovider); of pharmaceutical Chemistry Practice, calcium chemical Practice, device, 1996, Chatical Design of Drug, Chatical Design, Chapter et 1, and Chapter 1. Chapter

Thus, in one embodiment, the compound of formula (I) is a compound represented by:

the compound of formula (I) (or a salt or prodrug thereof) may be in a crystalline form, either as the free compound or as a solvate (e.g., hydrate), both forms being within the scope of the invention. Methods of solvation are well known in the art.

An "effective amount" is intended to mean an amount sufficient to effect treatment of AM L when formula (I) or a salt or prodrug thereof is administered to a subject in need of such treatment.

Treatment includes at least partially obtaining the desired effect, or delaying the onset or inhibiting the progression of AM L, or completely stopping or reversing the onset or progression of AM L.

In one embodiment, treatment is assessed by reducing the primary cell count (< 5%) and restoring normal blood cell count.

Under the FAB classification system, the AM L subtype can be the M0, M1, M2, M3, M4, M4eo, M5, M6 or M7 subtype AM L can also refer to AM L progenitor cells.

The compound of formula (I) (or a salt or prodrug thereof) may be administered to a patient at any stage of treatment, including induction, post-induction (or consolidation) and maintenance treatment stages, either as monotherapy or, more preferably, in combination with other induction, consolidation and/or maintenance therapies, including surgery, radiation or chemotherapy (e.g. antimetabolites, such as cytarabine (ara-C); anthracyclines, such as daunorubicin, doxorubicin or idarubicin; as well as other drugs, such as, such as 6-thioguanine, gemtuzumab ozogamicin (Mylotarg), midostaurin (Rydapt), enzipine (Idhifa), SCH-727965(Dinaciclib) and/or colony stimulating factors (e.g. G-CSF or GM-CSF).

In one embodiment, the compound of formula (I) (or a salt or prodrug thereof) is capable of specifically targeting or eliminating AM L cells (including AM L progenitor cells) during the induction and/or consolidation phase, resulting in complete remission and elimination.

The administration of the pharmaceutical combination of the invention may result not only in beneficial effects, e.g. additive or synergistic therapeutic effects, e.g. in terms of alleviating, delaying or inhibiting or ameliorating the symptoms of AM L, but also in therapeutic effects further surprising beneficial effects such other effects may include fewer adverse side effects, improved quality of life or reduced incidence compared to monotherapy applying only one of the pharmaceutically active ingredients used in the combination of the invention.

Another advantage of the present treatment regimen is that lower doses of the active ingredient of the compound of formula (I) can be used. Not only does the dosage need to be smaller, but the frequency of use can be reduced, which can reduce the incidence or severity of side effects.

In addition, the compounds of formula (I) may be used in combination with standard of care treatments. This allows standard of care treatment to be given at lower doses, thereby reducing the incidence or severity of side effects.

In addition, patients are typically monitored for AM L progression after treatment is initiated.

The term "administering" relates to administering a compound of formula (I) or a salt or prodrug thereof to a single patient. In combination therapy, if a treatment regimen is intended that includes agents wherein the agents need not be administered by the same route of administration or at the same time. Thus, one or more of the combined partner drugs administered may be administered together one after the other or separately, in one combined unit dosage form or in two separate unit dosage forms. The unit dosage form may also be a fixed combination, for example a pharmaceutical composition comprising two companion drugs.

For example, a method of treatment of AM L in accordance with the present invention may comprise (I) administration of a first combination partner in free or pharmaceutically acceptable salt form or prodrug form, (ii) administration of a second combination partner simultaneously or sequentially in any order, in jointly therapeutically effective amounts, preferably in synergistically effective amounts (e.g. in daily or intermittent doses corresponding to the amounts described herein), in free or pharmaceutically acceptable salt form or prodrug form.

As such, it will be understood that the combination of partners may be presented as a "kit of parts" for the treatment of AM L the kit may comprise a package wherein the combination partners are provided separately for co-administration with instructions for a particular therapy.

The effective dosage may vary depending on the particular compound or pharmaceutical composition used, the mode of administration, the condition being treated, the severity of the condition being treated. Thus, the dosage regimen will be selected in accordance with a variety of factors, including the route of administration and the renal and hepatic function of the patient. The effective amount of a single active required to alleviate, counter or arrest the progress of a disease can be readily determined and prescribed by the ordinarily skilled artisan.

The daily dosage will, of course, vary depending upon a variety of factors, such as the compound selected, the particular condition being treated, and the effect desired. However, in general, satisfactory results are obtained with the compounds of formula (I) administered at a daily dosage rate of about 0.05 to 18 mg/kg/day, e.g., 0.4 to 16 mg/kg/day, in single or divided doses. The compounds may be administered by any conventional route, in particular enterally or parenterally, for example in the form of injectable solutions or suspensions.

The compounds of formula (I) may be administered to a human in a daily dose range of 0.5 to 1000 mg. Suitable unit dosage forms for parenteral administration comprise from about 0.1 to 500mg of active ingredient, preferably 5-50 mg/day, more preferably 5-20 mg/day, most preferably about 7-12 mg/day, together with one or more pharmaceutically acceptable diluents or carriers.

Another benefit is that lower doses of the active ingredient may be used, for example, the dose need not only be often smaller but is also used less frequently, or may be used to reduce the incidence of side effects. This is in accordance with the desires and requirements of the patient to be treated.

The invention also relates to pharmaceutical compositions comprising a composition of formula (I) or a salt or prodrug thereof, e.g. comprising from about 0.1% to about 99.9%, including from about 1% to about 60%, of an active ingredient.

The composition, e.g., IV solution, may comprise any diluent or excipient. These include all conventional solvents, dispersion media, antibacterial agents, surfactants, isotonic agents and the like. It will be appreciated that the compositions of the invention may also comprise other supplementary physiologically active agents.

The carrier must be pharmaceutically "acceptable" in the sense of being compatible with the other ingredients of the composition and not deleterious to the subject. Compositions include those suitable for parenteral (i.e., intravenous) administration. The compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.

Compositions suitable for parenteral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain antioxidants, buffers, antimicrobials, and solutes which render the composition isotonic with the blood of the intended recipient. Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.

In one embodiment, the unit dosage compositions are those containing daily doses or units of the active ingredient, such as the daily sub-doses described above, or suitable fractions thereof.

It will be appreciated that the compositions of the invention may include other agents conventional in the art, in addition to the active ingredients specifically mentioned above, given the type of composition in question.

Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all such variations and modifications which fall within the spirit and scope of the invention. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features.

Certain embodiments of the present invention will now be described with reference to the following examples, which are for illustrative purposes only and are not intended to limit the general scope described above.

Examples of the invention

Synthetic schemes

Preparation of 2-bromo-7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran

Step 1-2-tert-butyldimethylsilyl-3- (tert-butyldimethylsilyloxymethylene) -6-methoxy-7-isopropoxybenzofuran (L arock coupling)

A suspension of 2-isopropoxy-3-methoxy-5-iodophenol (4.41mmol), 1- (tert-butyldimethylsilyl) -3- (tert-butyldimethylsiloxy) propyne (1.5g, 5.28mmol), lithium chloride (189mg, 4.45mmol) and sodium carbonate (2.34g, 22.08mmol) in anhydrous dimethylformamide (5m L) at 100 ℃ was deoxygenated by evacuation and backfilling with nitrogen 4 timesPalladium (135mg, 0.60mmol) was acidified and the reaction vessel was degassed twice with nitrogen then the reaction mixture was stirred at that temperature for 4 hours (tlc) and the solvent was removed by vacuum distillation the residue was dissolved in ethyl acetate (75m L), stirred well, filtered and treated with triethylamine (5m L) the solution was concentrated onto silica gel (10g) and purified by flash chromatography (silica gel, eluent hexane/diethyl ether/triethylamine; 95:5: 1%) to give the title compound as a yellow oil (1.45g, 96%);1H NMR(300MHz,CDCl3)7.24(d,1H,J=8.45Hz),6.88(d,1H,J=8.47Hz),4.80(s,2H,CH2),4.73(m,1H),3.88(s,3H,OMe),1.36(d,6H,J=6.17Hz),0.94(s,9H),0.92(s,9H),0.35(s,6H),0.12(s,6H)。

step 2: 2-tert-butyldimethylsilyl-3-formyl-6-methoxy-7-isopropoxybenzofuran

To a solution of 2-tert-butyldimethylsilyl-3- (tert-butyldimethylsilyloxymethylene) -6-methoxy-7-isopropoxybenzofuran (2.69mmol) in methanol (100m L) was added concentrated hydrochloric acid (200 μ L) and the reaction was stirred for 30 minutes (monitored by tlc), quenched with triethylamine (2m L) and the solvent was removed by distillation under vacuum the residue was dissolved in dichloromethane (20m L), washed with water (10m L), dried over magnesium sulfate, concentrated under vacuum and co-distilled with toluene (20m L) the crude product was dissolved in dry dichloromethane (4m L) and added to a stirred Collin reagent (chromium trioxide (1.01g), pyridine (1.65m L) in a solution of dry dichloromethane (30m L), the suspension was stirred for 10 minutes, filtered and the residue was washed with diethyl ether (20m L) the filtrate was concentrated onto silica (10g) and purified by flash chromatography (9 mg/hexane) to afford the title compound as a pale yellow oil, triethylamine (503%) as eluent: 48 mg/hexane;1H NMR(300MHz,CDCl3)10.25(s,1H,CHO),7.79(d,1H,J=8.45Hz),6.98(d,1H,J=8.46Hz),4.65(m,1H),3.89(s,3H,OMe),1.35(d,6H,J=6.17Hz),0.97(s,9H),0.45(s,6H)。

and step 3: 2-tert-butyldimethylsilyl-3- (3,4, 5-trimethoxybenzoyl) -6-methoxy-7-isopropoxybenzofuran

To a stirred solution of 3,4, 5-trimethoxyiodobenzene (377mg, 1.27mmol) in anhydrous tetrahydrofuran (1M L) under nitrogen at-78 ℃ was added n-butyllithium (795 μ L, 1.59mmol, 2M cyclohexane solution) and the reaction mixture stirred at this temperature for 40 minutes after which a solution of 2-tert-butyldimethylsilyl-3-formyl-6-methoxy-7-isopropoxybenzofuran (1.07mmol) in anhydrous tetrahydrofuran (1M L) was added dropwise by syringe pipette to the reaction mixture was stirred at-60 ℃ for 20 minutes then heated to 0 ℃, stirred for 10 minutes, quenched with saturated ammonium chloride solution (2M L) and diluted with ethyl acetate (20M L) the organic layer was washed with water (10M L), dried over magnesium sulfate and the solvent removed under vacuum to give a residue which was co-distilled with toluene 908mg of the crude product (908mg) dissolved in dry silica gel (10M) and treated with dichloromethane to obtain a yellow solid, after purification by flash chromatography, 1.5 mg of the title compound on silica gel (5 mg) and 1.5 mmol) to 5mg of silica gel, then treated with dichloromethane to obtain a yellow silica gel, 1.5 mg, 1.2 mg of the title compound (5 mmol) by syringe, 1mg) and washed with dichloromethane to obtain a yellow to obtain a mixture, followed by syringe, 1.498 to obtain a mixture by syringe, 1 to 5mg of benzoquinone by syringe, 1 to 10 mg, followed by syringe, 1 to obtain a mixture, 1 to 10 mg of toluene, 1 to obtain a mixture;1H NMR(300MHz,CDCl3)7.14(s,2H,benzoyl Hs),6.81(d,1H,J=8.64Hz),6.77(d,1H,J=8.64Hz)4.74(m,1H),3.93(s,3H,OMe),3.86(s,3H,OMe),3.78(s,6H,2x OMe),1.39(d,6H,J=6.14Hz),1.01(s,9H),0.26(s,6H)。

and 4, step 4: 2- (tert-butyldimethylsilyloxy) -7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran

To a stirred solution of 2- (tert-butyldimethylsilyloxy) -7-isopropoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxy-benzofuran (160mg, 0.31mmol) was added solid aluminum trichloride (83mg, 0.62mmol) under nitrogen atmosphere at room temperature, and the reaction mixture was stirred (monitored by tlc). the reaction was quenched with saturated ammonium chloride solution, extracted with dichloromethane and dried over magnesium sulfate, the solvent was removed by distillation, and the residue was dried by azeotropic removal of water with toluene. the crude product was dissolved in pyridine (2m L), acetic anhydride (1m L) was added, andthe reaction mixture was stirred at room temperature for 2 hours. The solvent was distilled under vacuum and the residue was loaded onto silica gel (1g) and purified by column chromatography (silica gel, eluent, hexane: diethyl ether; 80:20) (134mg, 84%);1H NMR(300MHz,CDCl3)7.14(s,2H, benzoyl Hs),6.98(d,1H, J ═ 8.72Hz),6.85(d,1H, J ═ 8.72Hz), 3.93(s,3H, OMe),3.86(s, 3H, OMe), 3.80(s,6H,2x OMe), 2.41(s,3H),0.99(s,9H),0.25(s, 6H).

And 5: 2-bromo-7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran

To a stirred solution of 2-tert-butyldimethylsilyl-7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran (120mg, 0.44mmol) in 1, 2-dichloroethane (1m L) was added bromine (12 μ L, 0.44mmol) dropwise at room temperature under a nitrogen atmosphere, and the reaction mixture was stirred at that temperature for 10 minutes after which the reaction was quenched with a saturated sodium thiosulfate solution, extracted with ethyl acetate (20m L), dried over magnesium sulfate, and the solvent was removed by vacuum distillation the crude product was purified by silica gel column chromatography (eluent: hexane: diethyl ether; 8:2-7:3) to give the title compound (91mg, 81%) as a colorless crystalline solid;1H NMR(300MHz,CDCl3)7.40(d,1H, J ═ 8.70Hz),7.14(s,2H, benzoyl-Hs), 6.98(d,1H, J ═ 8.75Hz),3.94(s,3H, OMe),3.89(s,3H, OMe),3.86(s,6H,2xOMe),2.43(s, 3H);13C NMR(75MHz,CDCl3)187.95(CO),167.71,152.75,149.54,147.49,142.59,131.92,131.80,123.91,121.84,119.89,117.72,109.89,106.92,60.69,56.61,56.00,20.09。

example 1

Preparation of 2-methyl-7-hydroxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxybenzofuran

Preparation A

To a stirred solution of 2-bromo 7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxy-benzofuran (20mg, 0.042mmol) and methylboronic acid (40mg, 0.67mmol) in 1, 4-dioxane (2m L) was added tetrakis-triphenylphosphine palladium (11mg, 0.01mmol) at 90 ℃, followed by sodium bicarbonate (40mg, 0.48mmol) in distilled water (0.5m L) after 5 minutes the reaction mixture became red after 2 hours (tlc), the reaction mixture was brought to room temperature and saturated ammonium chloride (2m L) was added and diluted with dichloromethane (20m L), the organic layer was separated, washed with water, dried over magnesium sulfate and the solvent was removed by vacuum distillation after which the residue was purified by PT L C (eluent dichloromethane/methanol, 1:1) to give the title compound (acetate cleaved during the reaction) as a fluffy white solid.

Preparation B (root-shore coupling)

To a stirred solution of zinc bromide (592mg, 2.63mmol) in anhydrous THF (1.5M L) at 0 ℃ was added methyllithium solution (1.6M in ether, 2.6M L, 4.15mmol), the reaction mixture was stirred for 2 hours solid 2-bromo-7-acetoxy-3- (3,4, 5-trimethoxybenzoyl) -6-methoxy-benzofuran (300mg, 0.63mmol) was added and the ether was removed under vacuum and dichlorobis (triphenylphosphine) palladium catalyst (21mg) and copper (I) iodide were added to the remaining suspension the reaction mixture was stirred for 36 hours at room temperature (monitored by tlc), quenched with saturated ammonium chloride solution and extracted with dichloromethane (10M L), dried with catalytic amounts of magnesium sulfate and the solvent was distilled off under vacuum and the product was purified by silica gel column (eluent hexane/ethyl acetate; 8:2) in methanol (106mg, 46%);1H NMR(300MHz,CDCl3)7.09(s,2H, benzoyl Hs),6.93(d,1H, J ═ 8.54Hz),6.83(d,1H, J ═ 8.56Hz),5.70(bs,1H, OH),3.93(s,3H, OMe),3.92(s,3H, OMe),3.83(s,6H,2x OMe),2.54(s,3H, 2-Me).

Example 2

Preparation of disodium 6-methoxy-2-methyl-3- (3,4, 5-trimethoxybenzoyl) benzofuran-7-yl phosphate

Step 1: dibenzyl 6-methoxy-2-methyl-3- (3,4, 5-trimethoxybenzoyl) benzofuran-7-yl phosphate:

to a mixture of 0.081g (0.22mmol) (7-hydroxy-6-methoxy-2-methylbenzofuran-3-yl) (3,4, 5-trimethoxyphenyl) methanone, 0.086g (0.261mmol) carbon tetrabromide, and 0.063m L (0.283mmol) dibenzylphosphite in 2.5m L anhydrous acetonitrile was added dropwise 0.046m L anhydrous triethylamine under nitrogen at 0 deg.C the resulting mixture was stirred at room temperature for 2h, then diluted with ethyl acetate to 20m L, washed with water and evaporated to dryness with anhydrous magnesium sulfate, filtered and evaporated under reduced pressure the residue was purified by flash column chromatography (dichloromethane/ethyl acetate, 9:1) to give the title compound as a colorless foam (0.13g, 94%);1H NMR(CDCl3)2.42(s,3H,Me-2),3.83(s,1H,OMe),3.93(s,3H,OMe),5.33(m,4H,CH2ph), 6.89(d, CH arene, J ═ 8.7Hz), 7.21(dd,1H, CH arene, J ═ 8.72 Hz; j ═ 1.2Hz), 7.08(s,2H, CH arene), 7.29-7.43 (m,10H, CH arene).

Step 2: 6-methoxy-2-methyl-3- (3,4, 5-trimethoxybenzoyl) benzofuran-7-sodium dihydrogen phosphate:

to a stirred solution of 0.122g (0.193mmol) of the product of step 1 in 1m L anhydrous acetonitrile under a nitrogen atmosphere at-5 ℃ was added 0.075m L (0.58mmol) bromotrimethylsilane, the resulting mixture was stirred at 0 ℃ for 1 hour then evaporated to dryness in vacuo, the residue was diluted to 5m L with anhydrous methanol and the pH of the solution was adjusted to about 10 by the addition of sodium methoxide after evaporation of the resulting mixture under reduced pressure, the solid residue was washed with anhydrous isopropanol (4 × 1.5.5 m L) and anhydrous ethanol (3 × 1.5.5 m L) and dried under vacuum to give 0.062g (65% yield) of the title compound as a colorless solid;1H NMR(D2O)2.37(s,3H,Me-2);3.76(s,6H,OMe);3.79(s,3H,OMe);3.82(s,3H,OMe);4.66(s,H2o); 6,93(d,1H, CH arene, J ═ 8.6 Hz); 7.04(d,1H, CH arene, J ═ 8.6 Hz); 7.10(s,2H, CH arene).

Biological data

Materials and methods

Tissue culture-cell lines

The cryopreserved cells were thawed in a water bath at 37 ℃ and the cells were suspendedThe solution was transferred to a 15M L tube containing 10M L of RPMI1640(Sigma) supplemented with 10% FBS (JRH Biosciences, catalog 12003, lot No. 5J0174), 100IU/M L penicillin (Sigma) and 100. mu.g/M L streptomycin (Sigma) (from now on referred to as RF10), or supplemented with 10% FBS (JRH Biosciences), 100IU/M L penicillin (Sigma) and L1-MEM (Sigma) of 100. mu.g/M8 streptomycin (Sigma) (from now on referred to as L MEM 10. MV 4; 11, L M13, H L, KG1 and ME1 in RF10, while OCI-AM L in α -MEM 10. the cells were cultured at 400g for 5 minutes, the supernatant was discarded, and resuspended in 5M 10 or RF 599% CO 2. the cells were resuspended in 52The culture is carried out in a humid incubator, and the culture is divided every 2 to 5 days to obtain 2 to 5 × 105Density of cells/m L.

Cytotoxicity and viability/proliferation assays

The cell line was expressed as 2 × 104Concentration of individual cells/well in 96-well plates (black, with clear flat bottom, Costar, 3603) inoculated in 50 μ L medium containing 0.1% DMSO and CellTox Green reagent (Promega, G8742), final dilution 1/500 for each dose tested, cells were plated in triplicate wells, drug was prepared to be 2-fold concentrated in 0.1% DMSO-containing medium immediately prior to use by reconstituting EX1 in DMSO to 10 mM., and 50 μ L was added to each well, fluorescence was measured at 24 and 48 hours using F L OUstar OPTIMA plate reader (BMG L abtech) (485 + 500 nm)Ex/520–530nmEm) After 48h cytotoxicity measurement, 20 μ L CellTiter-Blue reagent (Promega, G8080) was added to each well and the plates were incubated at 37 ℃ for 1h, followed by fluorescence measurement with F L OUstar OPTIMA plate reader (BMG L abtech) (560)Ex/590Em)。

Total ROS assay

1.25 × 105The cells were collected in 5M L tubes and washed with PBS the cell pellet was resuspended in 180 μ L PBS and 20 μ L of freshly prepared 5mM CM-H2DCFDA (Molecular Probes, C6827) solution was added to each tube as a positive control, tubes from which the cells were collected were treated with 160 μ L PBS to resuspend the cell pellet, 20 μ L of 10 μ M hydrogen peroxideT-butyl oxide (TBHP) and 20 μ L of 5mM CM-h2dcfda all tubes were incubated at 37 ℃ for 20 minutes, then cells were washed 3 times with PBS, resuspended in 300 μ L PBS and analyzed on a L srfortesta (bdbiosciences) Flow cytometer, results were analyzed using FCS Express4Flow Research Edition software (De Novo software).

Mitochondrial O2. cndot. -assay

1.25 × 105The cells were collected in 5M L tubes and washed with PBS the cell pellet was resuspended in 180 μ L PBS and 20 μ L of freshly prepared 5mM MitoSox (Molecular Probes, M36008) solution was added to each tube all tubes were incubated for 20 minutes at 37 ℃ and then the cells were washed 3 times with PBS the cells resuspended in 300 μ L PBS and analyzed on a L SRFortessa (BD biosciences) flow cytometer the results were analyzed using FCS Express 4FlowResearch Edition software (De Novo software).

Cell cycle assay

2 × 105The cells were collected in 5m L tubes and washed 3 times with PBS the cell pellet was suspended in 300 μ L cold PBS while vortexing, followed by 700 μ L of cold (-20 ℃) 100% ethanol was added dropwise to permeabilize the cells while vortexing, the cell suspension was incubated on ice for 30 minutes, centrifuged at 300g for 10 minutes, and then the cell pellet was resuspended in 100 μ L PI solution (0.1% Triton X-100, 100 μ g/m L RNAse A-Sigma without DNase and PBS at 40g/m L PI concentration). the cell suspension was incubated in the dark at 37 ℃ for 30 minutes, before analysis on L SRFortessa (BD biosciences) Flow cytometer, 200 μ L was added to each tube and the results were analyzed using FCS Express4Flow research PBS software (De Novo software).

Immunoblotting

Cells were collected (cell line 4 × 10)6,L-AMLs 7×106) And washed 3 times with cold PBS (centrifugation at 4 ℃). Lysis buffer was prepared with the recommended concentrations of NP40 cell lysis buffer (ThermoFisher Scientific), colomplete protease inhibitor (Roche, 046993116001), PhosStop (Roche, 04906837001) and Pefablock (Roche, 11585916001). After washing, the mixture is washedThe cells were resuspended in 50. mu. L lysis buffer, vortexed and incubated on ice for 30 minutes. the samples were vortexed every 10 minutes. after 30 minutes of incubation, the samples were centrifuged for 10 minutes at 14,000g, 4 ℃ and the supernatant was then transferred to tubes. according to the manufacturer's protocol, proteins were quantified using the DC Protein Assay kit (BioRad). The 50. mu.g of Cell line lysate or 20. mu.g of L-AM L lysate was loaded for SDS-PAGE. proteins were transferred from the gel to the PVDF membrane using a semi-dry transfer device (BioRad). The membrane was blocked with 5% skim milk containing 0.1% TBS-t for 1 hour at room temperature, then gently incubated with primary antibodies overnight at 4 ℃ in a gentle shaking. the antibodies were diluted in a dilution of anti-Noxa (Calbiochem, Cat. 1800-100 UG. batch 2875160) in 0.1% skim milk-t of 5% skim milk at a dilution ratio of 1: 500, anti-Heoling P catalog P-183-100 UG, batch 2875160) in a microtubes-5% skim milk-1. the dilution of 5% TBS-1. mu.1. Ab. As a microtubn. the results of the dilution of the anti-bovine serum Protein in a polyclonal Protein, the dilution of the ELISA, the anti-bovine serum, the ELISA, the monoclonal antibody, the ELISA, the monoclonal antibody was diluted with the monoclonal antibody.

Tissue culture-Primary sample

Thawing frozen mononuclear cells (MNC from BM or leukopheresis samples) in a water bath at 37 ℃ and transferring the Cell suspension to a 50M L tube 20% FBS (JRH biosciences), 50U/M L DNase I (Sigma), 100IU/M L penicillin (Sigma), 100 μ g/M L1 streptomycin (Sigma) and 2mM 2-glutamine (Sigma) added 20M L DM medium (Sigma, I3390), while vortexing the tube slowly (or mixing with hand) is added to the cells, centrifuging the cells at 400g for 5 minutes, discarding the supernatant, and washing the cells once with 20M 56 4 PBS, after washing, resuspending the cells in a tube containing 20% FBS (JSciences), 100IU L penicillin (Sigma), 100 μ g/M866 (Sigma) and 2mM PBS added to the Cell suspension at 20% FBS, JH Biotech) L, 365/M L after centrifugation at 400g speed for 5 minutes, 100 μ g/M L, after centrifugation at 365 g L, after centrifugation at 365 g L min, after centrifugation at 365 g L% of the supernatant, after centrifugation speed of the supernatant, and after centrifugation at L mM L, the centrifugation of the supernatant, the supernatant of the Cell suspension is transferred by centrifugation of the supernatant of the Cell suspension at L-L, the centrifugation of the supernatant, the centrifugation of the supernatant, the supernatant of the supernatant, the Cell suspension of the supernatant of the Cell suspension (PBS after centrifugation of the Cell suspension at L, the Cell suspension at L-3610-L, the Cell suspension at 3610-L, the Cell suspension at 3610-L, the centrifugation of the

(Selleckchem, S2858, batch 02) and 1 μ M UM729(Selleckchem, S7510, batch S751001). Viability and cell density were determined by trypan blue exclusion and cells were plated for each experiment as described in the results section.

Caspase-Glo3/7 assay

10 per sample/treatment4Individual cells (10 μ L) were aliquoted into triplicate wells of a 96-well plate (white wall OptiPlate 96, Perkin Elmer) containing 40 μ L complete medium 50 μ L Caspase-Glo3/7 reagent was added to each well, the plate was shaken for 30 seconds at 300-.

Viability (annexin V/7AAD) assay

2 × 10 collections in 5m L test tubes5Cells (200. mu. L) and washed 3 times with PBS cell pellet resuspended in 100. mu. L1 × binding buffer (B)D Biosciences, 51-66121E), the buffer contained 2 μ L Annexin V-APC antibody (BD Biosciences, catalog 550475, lot 6140584) and 2 μ L7 AAD solution (BD Biosciences, catalog 51-68981E, lot 5006667-1) and was incubated at room temperature in the dark for 30 minutes, 200 μ L PBS was added to the cell suspension before analysis on L srfortessa (BD Biosciences) flow cytometer, the results were analyzed using FCS Express 4FlowResearch Edition software (De Novo software).

L SC detection

Collection of 8 × 10 in 5m L test tubes5Cells (800 μ L) were fixed with 1.6% formaldehyde for 10 min at room temperature then samples were washed once with PBS and once with PBS/1% BSA the cell pellet was resuspended in 100 μ L PBS/1% BSA containing 5 μ L anti-CD 34-APC (Biosciences, clone 8G12, catalog 340441, batch 6183704), 5 μ L anti-CD 38-PECy7(BD Biosciences, 335790, batch 3353807) and 5 μ L anti-GPR 56-PE (Biolegend, 358204, batch B208208208), incubated 1h at room temperature protected from light and finally the cells were washed twice with PBS and then resuspended in 200 μ L PBS 353 for analysis on a L srfortestassa (BD Biosciences) Flow cytometer FCS Express4Flow Research software (desvo software).

Colony assay

Method H4034(STEMCE LL Technologies) at 1.5m L was aliquoted into 5m L tubes (one tube/treatment/sample) and supplemented with 20ng/m L of hI L-6 (Peprotech, 200-06). EX1 and OXi4503 dilutions were added to the tubes, respectively, to final concentrations of 100nM and 500 nM. DMSO was used as a blank control.samples were added to each tube to reach final cell densities L-AM L6 and L-AM L419 to 2 × 104Cells/well, AM L136 and AM L480 of 3 × 103Cells/well. The tubes were shaken to mix the drug and cells with Methocult and incubated at 37 ℃ for 10 minutes to remove air bubbles. The samples were then plated in 24-well plates and 4 wells/treatment/sample in 8 wells in the center of the plate. The holes in the rim are filled with sterile water to avoid drying. Plates were incubated at 37 ℃ for 13 days before counting colonies.

MTS proliferation assay

Cell lines (MV 4; 11 and H L-60) were tested at 1 × 104Concentration of individual cells/well was seeded in 50u L complete growth medium in 96-well plates for each test dose, cells were plated in triplicate wells EX1 (in DMSO), Oxi4503(endo therm, batch # EN09013, in water), CA4P (seleckchem, Cat # S7204, in water), and Plinabulin (seleckchem, Cat # S1176, in DMSO), immediately prior to use, the drug was prepared to 2-fold concentration in the medium in 10 mM. as indicated in appropriate diluents, and 50u L (0.1% DMSO or water, final concentration) was added to each well, respectively, after 72 hours, 20 μ L l titer 96 aqua One Solution reagent (Promega catalog No. G3581) was added to each well and incubated for 2 hours at 37 ℃ and then read on a thermomechanical 96 absorbance in a microplate 384.

Dose response of AM L cell line to example 1(EX 1)

To test the cytotoxic and antiproliferative effects of EX1 on the AM L cell line, six different cell lines (table 1) were thawed and cultured in respective media, and then treated as described below.

TABLE 1 detailed characteristics of the AM L cell line used in this study

The first experiment was performed with H L60 and MO L M13 cell lines, expanding cells, when sufficient in number (survival rate higher than 90%), and changing them to 2 × 10%4The amount of individual cells/well was seeded in 50. mu. L media each containing 0.1% DMSO and CellTox Green reagent the dose range tested was 1. mu.M, then 1/10 dilutions were performed sequentially until 10-5nM EX1, or solvent only. Plates were incubated and fluorescence (indicative of cytotoxicity) was measured at 24h and 48 h. After 48 hours reading cytotoxicity, CellTiter-Blue reagent was added to each well. The reading of this assay is based on the metabolic activity of the cells and serves as a surrogate marker for proliferation and/or cell viability.

Following the same protocol as above, the next experiment was performed with the cell lines ME1, OCI-AM L, MV 4; 11 and KG1, as can be seen from the previous experimental results, the non-linear curve fit was very steep between 1nM and 0.1nM (FIGS. 1A and B), therefore, the drug dose was adjusted for this and subsequent experiments to one that was prepared for the H L and MO L M13 experiments a week ago for 1 μ M, 100nM, 10nM, 1nM, 0.75nM, 0.5nM, 0.25nM, 0.1nM, 0.01nM and 0.829001 drug stock, stored at-20 ℃ C. the cytotoxicity and proliferation (metabolic assay) were measured as above, finally, additional experiments were performed for MV 4; 11 and MO L M13, fresh samples were aliquoted to confirm the results obtained IC50 values as shown in FIGS. 1A and B, and the IC50 data were pooled in the case of multiple experiments to determine IC 50.

As shown in FIGS. 1A and B, all cell lines showed sensitivity to EX1, with very low IC50 doses, ranging from-0.2 nM to 1.3 nM. cytotoxicity and proliferation assays showing that the cells were divided into two groups, the most sensitive cell lines (H L60, MO L M13 and MV 4; 11) reached the highest cytotoxicity and showed the greatest inhibition of proliferation, the less sensitive cell lines (ME1, OCI-AM L3 and KG1) showed lower cytotoxicity and also kept above background readings in the CellTiter assay, consistent with some cell viability at higher drug concentrations.

2. Study of the mechanism by which EX1 targets AM L cells

Once it was determined that this group of AM L cell lines was highly sensitive to EX1, the underlying mechanism of this reaction was investigated, for which 10 cells were treated6Cells/m L were seeded and treated with vehicle (DMSO) or a series of doses of EX 1(0.1nM, 1nM, 10nM and 100nM) for 6h, 8h and 24h, respectively, cells were harvested for detection of ROS and superoxide anion (O2. cndot. -) levels at 6h and 24h, proteins were extracted at 8h and 24h, and cell cycle analysis at 24 h.

2.1 Generation of Reactive Oxygen Species (ROS) by cells treated with EX1

As reported in the literature, KG1 cells treated with compstatin (combretastatin) -a4-P and OXi4500 (compstatin-a 1) showed increased total ROS levels (Petit et al. blood 2008; madrambayan et al. blood2010), which were measured in 6 cell lines after treatment with a series of doses of EX1 for 6h and 24 h. Total ROS levels were measured by flow cytometry using the fluorescent dye CM-H2DCFDA, at a final concentration of 5. mu.M. The level of mitochondrial superoxide anion (O2. cndot. -) was measured by flow cytometry using MitoSox, again at a final concentration of 5. mu.M.

As shown in figure 2A, the dose response of total ROS in OCI-AM L cells was reduced 24 hours after treatment, but none of the other cell lines showed any change in total ROS levels (data not shown). on the other hand, mitochondrial O2-levels were changed in most cell lines, but there were two different patterns.mv 4; 11 and MO L M13 both showed a dose response to the drug, with an increase in mitochondrial O2-levels observed only at 24 hours, which was more pronounced in MO L M13 cells (figure 2B). in contrast, H L and ME 84 had a slightly reduced mitochondrial O2-levels at 6 hours (data not shown), while the observed effect was greater in ME1 cells at 24 hours (figure 2B). although OCI-AM L had a slight decrease in mitochondrial O2-levels at 6 hours, but an increase in O2-levels at 24 hours (figure 2B) appeared to have no detectable change in mitochondrial O3638-levels at 24 hours (figure 2B).

2.2 cell cycle analysis after treatment of AM L cell line with EX1

Petit and his collaborators also showed that combretastatin A4-P induced G2/M cell cycle arrest in KG1 cells (Petit et al, Blood 2008). Thus, as described above, cell cycle analysis was performed in cell lines by PI staining after 24 hours of treatment with a series of doses of EX 1.

As shown in FIG. 3, there are two patterns of cell cycle distribution after 24 hours of treatment with EX1 MV4, 11 and MO L M13 cells undergo apoptosis, as evidenced by an increase in the sub-G0 population furthermore these cell lines show a dose-dependent decrease in the percentage of G2/M and G1 cells, for the remaining cell lines H L60, KG1, OCI-AM L3 and ME 1a second pattern was observed, the data for all these cell lines are consistent with G2/M arrest in response to EX1 KG1 and H L60 also show a significant increase in the sub-G0 cell population consistent with induction of apoptosis.

2.3 activation of the pro-apoptotic signals

EX1 has been shown to induce apoptosis in chronic lymphocytic leukemia (C LL) cells by phosphorylation of JNK (P-JNK) and induction of Noxa (Bates et al, Cancer biol. Ther.2016.) thus, as described above, treatment of cells with a range of doses of EX1 prior to protein extraction for 8h and 24 h. Western blot analysis showed that at 8h all cell lines induced P-JNK KThr183/Tyr185 (FIG. 4) strongly in EX1 at 10hM and 100nM, on the other hand, induction of Noxa could only be detected in OCI-AM L3 and ME1, with both 10nM and 100nM of drug and 8h post-treatment Noxa readily detected in control lysates prepared from HEK293 cells.

Taken together, these results indicate that even though EX1 consistently induces mechanisms of JNK activation, apoptosis or cell cycle arrest in the AM L cell line, may still be cell type dependent, MV 4; 11 and MO L M13 show similar responses to EX1 based on cytotoxicity, mitochondrial O2-and cell cycle, while the other group (KG1, ME1 and OCI-AM L3) shows lower levels of cytotoxic responses and cell cycle distribution.

3. Targeting primary AM L cells by EX1

For these experiments, cryopreserved samples of de-characterized AM L (monocytes, MNC) were retrieved from SACRB bio-banks according to ethical approval by the royal aldleyder human research ethical committee (RAH HREC) (R20170220).

AM L bone marrow specimen

Experiments were performed using three Healthy Control (HC) BMMNC samples (HC61, HC68, and HC71) and other AM L samples (MNCs isolated from bone marrow diagnostic samples, table 2.) as described in materials and methods, cells were thawed, recovered overnight in the described media, and checked the viability and cell number the next day (by trypan blue exclusion), overnight recovery and viability check, after which dead cells were removed by density gradient centrifugation with L ymphoprep to purify two samples (AM L235 and AM L342). after purification, the viable cells recovered from AM L342 after purification were insufficient for the experiments and therefore not used6Individual cells/m L (500. mu. L/well, duplicate wells) were plated in 24-well plates and AM L s at 106Plates of individual cells/m L were plated in 6-well plates at 2m L/well/dose since BM AM L samples had fewer cells available than L-AM L (leukapheresis), these samples were not plated to prepare protein lysates, but were used only for antibody staining and flow cytometry analysis (annexin v/7AAD, and CD34, CD38, and GPR56 staining to detect leukemic stem cell L SC populations).

TABLE 2 characterization of the AM L main sample used in this study

PB peripheral blood, BM bone marrow, M LL, possible M LL changes, t-AM L, AM L associated with therapy.

FeasibilityAfter 24 hours of treatment, 10 μ L cells were used in the Caspase-Glo3/7 assay as before, again little to no Caspase (cysteine-containing aspartate proteolytic enzyme) activation was observed (fig. 5A). only AM L was shown to increase Caspase activity in cells treated with EX1, but already high activity in DMSO, which means very low viability of the sample, based on this result, when the Caspase-Glo3/7 assay was performed again, the sample was cultured for one day again at 48 hours, Caspase activity was increased in all AM L samples treated with EX1 compared to DMSO (fig. 5B). then 200 μ L cells were collected from all samples for Annexin V/7AAD staining, and 800 μ L AM L culture was used for CD34, CD38 and GPR 25 staining, and the remaining cells were cultured for the same day for Annexin the collection of cells for Annexin V/7AAD staining, and the remaining cells were also shown to be stable in response to the previous experiments (fig. 5C) at 72C, showing a decrease in survival response to the time of the sample after the wash, but the wash, development of the wash, no Caspase activity was observed at 48 hours (fig. 5C, 3C, 5C, showing a decrease in the wash, 5C, showing a decrease in response to the wash, showing a decrease in the wash, wash, wash.

LSC analysisFirst, viable cells in AM L BMMNC samples were gated and then the proportion of GPR56+ cells was determined according to the report by Pabst and its co-workers (Pabst et al, Blood 2016), in CD34highIn the AM L sample, GPR56+ population containing L SC was present within CD34+ cells, whereas in CD34lowThus, the CD34+/GPR56+ gate of AM 390235 and the GPR56+/CD34+ and GPR56+/CD 34-gates of AM L and AM L estimated that subpopulations containing L SC (fig. 6A) will be consistent with drugs targeting the vital compartment containing L SC in response to reduction of these subpopulations of EX1 as shown in fig. 6B, flow cytometry analysis indicated that EX1 affects this population at different rates in a sample-dependent manner.

Primary cell colony assayFor L-AM L, a small number of colonies and high background single cells (i.e., cells that survived in semi-solid medium but did not form colonies) were observed in control treated wells, whereas no colonies or background cells were observed in wells using either dose of EX1 (FIG. 7A), treatment of the sample with 100nM OXi4503 reduced background cells and reduced colony numbers, whereas treatment with 500nM completely blocked colony formation and background of live cells (FIG. 7A), L-AM 36419 also formed a small number of colonies in DMSO treated wells, background cells were reduced, whereas treatment with both drugs (all doses) completely blocked colony formation and background formation of the sample (FIG. 7B).

Western blot analysisAfter 48 hours of treatment, two of the AM L samples (L-AM L6 and L-AM L419) were prepared for protein lysate for samples L-AM L419, insufficient protein was recovered and no Western blot analysis could be performed for the L-AM L6 cells, using cells that had been tested in cell line experimentsThe evidence specific antibodies determined P-JNK KThr183/Tyr185 and Noxa expression (FIG. 4). A slight increase in P-JNK KThr183/Tyr185 was observed for cells treated with EX1 at 500nM, whereas no significant change was observed with OXi4503 (FIG. 8). Noxa expression was detectable, but very low in this sample, and no change was observed in both drug treatments. The important point to consider here is that the protein yield of this sample is very low, loading only 20. mu.g of protein for SDS-PAGE and 50. mu.g of protein for cell line samples.

Comparison of EX1 with other tubulin polymerization inhibitors

To compare the efficacy of EX1 with other tubulin polymerization inhibitors, MTS proliferation assays were performed on MV 4; 11 and H L-60 AM L cell lines (Table 1). cells were expanded and, when sufficient in number (and greater than 90% viability), were treated at 1 × 104The concentration of individual cells/well was seeded in 100 μ L complete growth medium containing a final concentration of 0.1% DMSO or water in 96-well plates EX1, Oxi4503(endo therm, batch # EN09013 in water), CA4P (Selleckchem, catalog number S7204 in water), and Plinabulin (Selleckchem, catalog number S1176 in DMSO) were tested and diluted from 0.00001nM to 1000nM in serial 1/10 cells were cultured with the compounds for 72 hours, then viability was measured by addition of CellTiter 96AQueous One Solution reagent (promegat # G3581) and absorbance was measured at 492nM ic50 was calculated using the absolute IC50 method in GraphPad Prism 6 as shown in figure 9, EX1 had the lowest IC50 in two AM L cell lines, indicating that it is more effective than other tested tubulin polymerization inhibitors.

31页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:用于治疗与纤毛病相关联的疾病的方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!