A Conformational Restriction Strategy for the Identification of a Highly Selective Pyrimido-pyrrolo-oxazine mTOR Inhibitor
- Chiara BorsariChiara BorsariDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Chiara Borsari
- ,
- Denise RageotDenise RageotDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Denise Rageot
- ,
- Alix Dall’AsenAlix Dall’AsenPIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, SwitzerlandMore by Alix Dall’Asen
- ,
- Thomas BohnackerThomas BohnackerDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Thomas Bohnacker
- ,
- Anna MeloneAnna MeloneDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Anna Melone
- ,
- Alexander M. SeleAlexander M. SeleDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Alexander M. Sele
- ,
- Eileen JacksonEileen JacksonDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Eileen Jackson
- ,
- Jean-Baptiste LangloisJean-Baptiste LangloisDepartment of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Jean-Baptiste Langlois
- ,
- Florent BeaufilsFlorent BeaufilsPIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, SwitzerlandMore by Florent Beaufils
- ,
- Paul HebeisenPaul HebeisenPIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, SwitzerlandMore by Paul Hebeisen
- ,
- Doriano FabbroDoriano FabbroPIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, SwitzerlandMore by Doriano Fabbro
- ,
- Petra HillmannPetra HillmannPIQUR Therapeutics AG, Hochbergerstrasse 60, 4057 Basel, SwitzerlandMore by Petra Hillmann
- , and
- Matthias P. Wymann*Matthias P. Wymann*E-mail: [email protected]. Phone: +41 61 207 5046. Fax: +41 61 207 3566.Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, SwitzerlandMore by Matthias P. Wymann
Abstract

The mechanistic target of rapamycin (mTOR) plays a pivotal role in growth and tumor progression and is an attractive target for cancer treatment. ATP-competitive mTOR kinase inhibitors (TORKi) have the potential to overcome limitations of rapamycin derivatives in a wide range of malignancies. Herein, we exploit a conformational restriction approach to explore a novel chemical space for the generation of TORKi. Structure–activity relationship (SAR) studies led to the identification of compound 12b with a ∼450-fold selectivity for mTOR over class I PI3K isoforms. Pharmacokinetic studies in male Sprague Dawley rats highlighted a good exposure after oral dosing and a minimum brain penetration. CYP450 reactive phenotyping pointed out the high metabolic stability of 12b. These results identify the tricyclic pyrimido-pyrrolo-oxazine moiety as a novel scaffold for the development of highly selective mTOR inhibitors for cancer treatment.
Introduction
Figure 1

Figure 1. Chemical structures of rapamycin and rapalogs and a selection of ATP-competitive mTOR kinase inhibitor (TORKi) compounds.
Results and Discussion
Rigidification Strategy
Figure 2

Figure 2. Strategy for the development of mTOR selective inhibitors starting from PQR309 (1): rigidification strategy (red dotted lines) and removal of trifluoromethyl group from the 2-aminopyridine moiety (blue).

cellular assay, IC50 [nM]a | in vitro binding assay, Ki [nM]b | |||||
---|---|---|---|---|---|---|
compd | pPKB S473 | pS6 S235/236 | p110α | mTOR | selectivity Ki(p110α)/Ki(mTOR) | clogPc |
PQR309 (1) | 139 | 205 | 17 | 62 | 0.27 | 3.11 |
2a | 194 | 164 | 76 | 69 | 1.1 | 2.65 |
2b | 118 | 96 | 432 | 24 | 18.3 | 2.65 |
2c | 1871 | 1790 | 867 | 812 | 1.1 | 2.65 |
2d | 1560 | 1857 | 541 | 1279 | 0.4 | 2.65 |
PKB phosphorylation on Ser473 and ribosomal S6 phosphorylation on Ser235/236 were analyzed in A2058 cells exposed to the indicated inhibitors and subsequent detection of phosphoproteins in an in-cell Western assay. Each experiment was performed with n = 2. The log IC50 values and standard errors are reported in Table S7 in the Supporting Information.
Compounds were tested for the in vitro binding to the ATP-binding site of p110α and mTOR using a commercially available time-resolved FRET (TR-FRET) displacement assay (LanthaScreen). Each experiment was performed with n = 2. IC50 values and log IC50 values and standard errors are reported in Table S7 in the Supporting Information.
Marvin/JChem 16.10.17 was used for calculation of log P (partition coefficient) values.
Elucidation of Binding Modes to PI3K and mTOR

PKB phosphorylation on Ser473 and ribosomal S6 phosphorylation on Ser235/236 were analyzed in A2058 cells exposed to the indicated inhibitors and subsequent detection of phosphoproteins in an in-cell Western assay. Each experiment was performed with n = 2. The log IC50 values and standard errors are reported in Table S8.
Compounds were tested for the in vitro binding to the ATP-binding site of p110α and mTOR using a commercially available time-resolved FRET (TR-FRET) displacement assay (LanthaScreen). Each experiment was performed with n = 2. IC50 values, logIC50 values, and standard errors are reported in Table S8.
Figure 3

Figure 3. (A) Docking of compound 3a (plum) into PI3Kγ (gray) starting from PDB code 5JHB (see ref (32)). Structural water molecules are shown in red, and water-mediated H-bonds are depicted as dashed black lines. (B) Docking of compound 2a (gold) and (C) compound 2b (green) into mTOR (turquoise) starting from PDB code 4JT6. The important features for mTOR selectivity are depicted in a ball and stick representation. (D) Docking of compound 2a (gold) and (E) compound 2b (green) into PI3Kα (gray) starting from PDB code 3ZIM. The exit vector from the restricted morpholine oxygen is shown as a black arrow.
Scheme 1

Scheme 1. c
aPrepared according to ref (32).
bPrepared according to procedure vii. After the reaction, the two regioisomers (26 and 32) were separated by column chromatography.
c(A) Reagents and conditions: (i) (1) benzaldehyde, 2 M NaOH, rt, 30 min; (2) NaBH4, 5 °C → rt, 1 h; (ii) (1) chloroacetyl chloride, K2CO3, THF/H2O, 0 °C, 1 h; (2) NaOH, 5 °C, 2 h; (iii) borane–dimethyl sulfide complex, Et3N, THF, 0 °C → 65 °C, 5 h; (iv) Pd/C, H2, 2.8 bar, 48 h; (v) thionyl chloride, imidazole, DCM, −5 °C → rt → 0 °C, 2 h; (vi) ruthenium(IV) oxide hydrate, NaIO4, rt, o/n. (B) Reagents and conditions: (vii) morpholine derivative (Mn–H), DIPEA, DCM, 0 °C → rt, o/n; (viii) (1) n-BuLi, CuI, −78 °C → rt, o/n; (2) HCl conc, MeOH, 45 °C, 4–6 h; (3) NaOH, H2O, rt, 1–16 h; (ix) (1) boronic ester 38 or 41, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 2–16 h; (2) HCl, dioxane/H2O, 60 °C, 3–16 h (for 2a, 2b, 2c, 2d, 6a, 6b, 7a, 7b, 8a, 8b, 9a, 9b, 10a, 10b, and 14b); (x) 2-aminopyridine-5-boronic acid pinacol ester, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C (for 11b), o/n; (xi) boronic ester 39, Pd(dppf)Cl2 (cat.), CsCO3, THF, Δ, o/n (for 12b); (xii) (1) boronic ester generated in situ, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 3–3.5 h; (2) HCl, 80 °C, o/n (for 13b and 15b); (xiii) (1) boronic ester generated in situ, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 2–16 h (isolated intermediates 18b and 19b); (2) 18b or 19b, TFA, DCM, 0 °C → rt, 1–3 h (for 16b and 17b).
Chemistry
Determination of Cellular Potency and PI3K vs mTOR Kinase Activities

PKB phosphorylation on Ser473 and ribosomal S6 phosphorylation on Ser235/236 were analyzed in A2058 cells exposed to the indicated inhibitors and subsequent detection of phosphoproteins in an in-cell Western assay. Each experiment was performed with n = 2. The log IC50 values and standard errors are reported in Table S7 in the Supporting Information.
Compounds were tested for the in vitro binding to the ATP-binding site of p110α and mTOR using a commercially available time-resolved FRET (TR-FRET) displacement assay (LanthaScreen). Each experiment was performed with n = 2. IC50 values, log IC50 values, and standard errors are reported in Table S7 in the Supporting Information.
Marvin/JChem 16.10.17 was used for calculation of log P (partition coefficient) values.

PKB phosphorylation on Ser473 and ribosomal S6 phosphorylation on Ser235/236 were analyzed in A2058 cells exposed to the indicated inhibitors and subsequent detection of phosphoproteins in an in-cell Western assay. Each experiment was performed with n = 2. The log IC50 values and standard errors are reported in Table S7 in the Supporting Information. *PQR620 data are from ref (31) for comparison.
Compounds were tested for the in vitro binding to the ATP-binding site of p110α and mTOR using a commercially available time-resolved FRET (TR-FRET) displacement assay (LanthaScreen). Each experiment was performed with n = 2. IC50 values, log IC50 values, and standard errors are reported in Table S7 in the Supporting Information.
Marvin/JChem 16.10.17 was used for calculation of log P (partition coefficient) values.
Pharmacological Parameters
Figure 4

Figure 4. Plasma and brain concentration of (A) compound 7b and (B) compound 12b after po dosing at 5 mg/kg in male Sprague Dawley rats. Stability of compound 11b (5 μM) with primary hepatocytes from (C) mice (green) and rats (turquoise) and (D) dogs (red) and humans (black) (n = 2). All values are the mean ± SEM. Error bars are not shown when smaller than the symbols.
CYP450 Reactive Phenotyping for Compounds 7b, 11b, and 12b
remaining test item with cofactors | % remaining in corresponding negative control without cofactors | ||||
---|---|---|---|---|---|
test item | mean % | SD | mean | SD | mean (%)corrb |
CYP1A1 | |||||
7b | 44 | 2.0 | 109 | 3.7 | 35 |
11b | 51 | 0.3 | 93 | 2.9 | 58 |
12b | 77 | 0.08 | 96 | 4.5 | 81 |
CYP1A2 | |||||
7b | 98 | 0.6 | 94 | 0.1 | 104 |
11b | 73 | 0.4 | 91 | 2.4 | 83 |
12b | 92 | 0.2 | 91 | 0.8 | 101 |
Compounds remaining after 60 min of incubation with Supersomes at 25 pmol/mL (n = 2), calculation based on absolute amounts (nM).
mean (%)corr = (100 – % remaining negative control) + % remaining sample.
Figure 5

Figure 5. Pie chart showing the percentage of compound 11b and its metabolites after 60 min of incubation with human recombinant (A) CYP1A1 and (B) CYP1A2. (C) Major metabolites observed upon incubation of 11b with CYP1A1.
Enzymatic Profiling and Determination of Selectivity
inhibitor binding constant,aKd [nM] | ||||||||
---|---|---|---|---|---|---|---|---|
compd | mTOR | PI3Kα | PI3Kβ | PI3Kδ | PI3Kγ | PI4Kβ | VPS34 | most sensitive PI3K/mTOR,c fold selectivity |
7b | 34 | 610 | 4100 | 6200 | 8700 | >30000 | 2100 | 17.9 |
11b | 14 | 120 | 1400 | 1900 | 1800 | >30000 | 980 | 8.6 |
12b | 3.5 | 1600 | 7500 | 12000 | 11000 | >30000 | 3200 | 457 |
CC223 (55)b | 28 | 2300 | 18000 | 6200 | 7100 | 39 | 2500 | >80× |
INK128 (56)b | 0.092 | 15 | 81 | 30 | 3.7 | nd | 8200 | >40× |
AZD2014 (57)b | 0.14 | 33 | 3300 | 1500 | 8400 | >30000 | 23000 | >230× |
PQR620 (58) | 0.27 | 1000 | 22000 | 23000 | 18000 | >30000 | 2750 | >3700× |
Dissociation constants (Kd) were determined using ScanMax technology (DiscoveRx) with 11-point 3-fold serial dilutions of the indicated compounds. Kd is the mean value from experiments performed in duplicate and was calculated from standard dose–response curves using the Hill equation. nd = not determined.
Dissociation constants (Kd) of CC223 (55), INK128 (56), AZD2014 (57), and PQR620 (58) are from ref (31).
Fold selectivity: ratio of Kd of the most sensitive class I PI3K isoform (displayed in bold type) over Kd for mTOR.
Conclusion
Experimental Section
General Information
General Procedure 1
General Procedure 2
General Procedure 3
General Procedure 4
General Procedure 5
Step 1
Step 2
General Procedure 6
(S)-5-(4-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (2a)
(R)-5-(4-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (2b)
(S)-5-(2-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-4-yl)pyridin-2-amine (2c)
(R)-5-(2-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-4-yl)pyridin-2-amine (2d)
(S)-5-(4-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (3a)
(R)-5-(4-Morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (3b)
(S)-5-(4-Morpholino-5,5a,6,7,8,9-hexahydropyrimido[5,4-b]indolizin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (4a)
(R)-5-(4-Morpholino-5,5a,6,7,8,9-hexahydropyrimido[5,4-b]indolizin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (4b)
(S)-5-(4-(Piperidin-1-yl)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (5a)
(R)-5-(4-(Piperidin-1-yl)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (5b)
5-((S)-4-((S)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (6a)
5-((R)-4-((S)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (6b)
5-((S)-4-((R)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (7a)
5-((R)-4-((R)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (7b)
5-[(9S)-6-{8-Oxa-3-azabicyclo[3.2.1]octan-3-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyridin-2-amine (8a)
5-[(9R)-6-{8-Oxa-3-azabicyclo[3.2.1]octan-3-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyridin-2-amine (8b)
5-[(9S)-6-{3-Oxa-8-azabicyclo[3.2.1]octan-8-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyridin-2-amine (9a)
5-[(9R)-6-{3-Oxa-8-azabicyclo[3.2.1]octan-8-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyridin-2-amine (9b)
(S)-5-(4-(3,3-Dimethylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (10a)
(R)-5-(4-(3,3-Dimethylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (10b)
5-((R)-4-((R)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyrimidin-2-amine (11b)
5-((R)-4-((R)-3-Methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyrazin-2-amine (12b)
4-Methyl-5-((R)-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (13b)
3-Methyl-5-((R)-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (14b)
4-Methoxy-5-((R)-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (15b)
4-(Dimethoxymethyl)-5-((R)-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyridin-2-amine (16b)
4-(Dimethoxymethyl)-5-((R)-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazin-2-yl)pyrimidin-2-amine (17b)
tert-Butyl N-[(tert-Butoxy)carbonyl]-N-[4-(dimethoxymethyl)-5-[(9R)-6-[(3R)-3-methylmorpholin-4-yl]-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyridin-2-yl]carbamate (18b)
tert-Butyl N-[(tert-Butoxy)carbonyl]-N-[4-(dimethoxymethyl)-5-[(9R)-6-[(3R)-3-methylmorpholin-4-yl]-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-trien-4-yl]pyrimidin-2-yl]carbamate (19b)
(S)-2-Chloro-4-morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (20a)
(R)-2-Chloro-4-morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (20b)
(S)-4-Chloro-2-morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (20c)
(R)-4-Chloro-2-morpholino-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (20d)
(S)-2-Chloro-4-((S)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (21a)
(R)-2-Chloro-4-((S)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (21b)
(S)-2-Chloro-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (22a)
(R)-2-Chloro-4-((R)-3-methylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (22b)
(9S)-4-Chloro-6-{8-oxa-3-azabicyclo[3.2.1]octan-3-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-triene (23a)
(9R)-4-Chloro-6-{8-oxa-3-azabicyclo[3.2.1]octan-3-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-triene (23b)
(9S)-4-Chloro-6-{3-oxa-8-azabicyclo[3.2.1]octan-8-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-triene (24a)
(9S)-4-Chloro-6-{3-oxa-8-azabicyclo[3.2.1]octan-8-yl}-11-oxa-1,3,5-triazatricyclo[7.4.0.02,7]trideca-2(7),3,5-triene (24b)
(S)-2-Chloro-4-(3,3-dimethylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (25a)
(R)-2-Chloro-4-(3,3-dimethylmorpholino)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (25b)
4-(2,6-Dichloropyrimidin-4-yl)morpholine (26)
(S)-4-(2,6-Dichloropyrimidin-4-yl)-3-methylmorpholine (27)
(R)-4-(2,6-Dichloropyrimidin-4-yl)-3-methylmorpholine (28)
3-(2,6-Dichloropyrimidin-4-yl)-8-oxa-3-azabicyclo[3.2.1]octane (29)
8-(2,6-Dichloropyrimidin-4-yl)-3-oxa-8-azabicyclo[3.2.1]octane (30)
4-(2,6-Dichloropyrimidin-4-yl)-3,3-dimethylmorpholine (31)
4-(4,6-Dichloropyrimidin-2-yl)morpholine (32)
(S)-Tetrahydro-3H-[1,2,3]oxathiazolo[4,3-c][1,4]oxazine 1,1-Dioxide (33a)
(R)-Tetrahydro-3H-[1,2,3]oxathiazolo[4,3-c][1,4]oxazine 1,1-Dioxide (33b)
(S)-Morpholin-3-ylmethanol (34a)
(R)-Morpholin-3-ylmethanol (34b)
(S)-(4-Benzylmorpholin-3-yl)methanol (35a)
(R)-(4-Benzylmorpholin-3-yl)methanol (35b)
(S)-4-Benzyl-5-oxomorpholine-3-carboxylic Acid (36a)
(R)-4-Benzyl-5-oxomorpholine-3-carboxylic Acid (36b)
N-Benzyl-(S)-serine (37a)
N-Benzyl-(R)-serine (37b)
(E)-N,N-Dimethyl-N′-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)formimidamide (38)
5-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)pyrazin-2-amine (39)
(E)-N′-(5-Bromo-4-methylpyridin-2-yl)-N,N-dimethylformimidamide (40)
(E)-N,N-Dimethyl-N′-(3-methyl-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)pyridin-2-yl)formimidamide (41)
tert-Butyl (5-Bromo-4-methoxypyridin-2-yl)carbamate (42)
5-Bromo-4-(dimethoxymethyl)pyridin-2-amine (43)
tert-Butyl N-[5-Bromo-4-(dimethoxymethyl)pyridin-2-yl]-N-[(tert-butoxy)carbonyl]carbamate (44) and tert-Butyl N-[5-Bromo-4-(dimethoxymethyl)pyridin-2-yl]carbamate (45)
(E)-4-(Dimethylamino)-1,1-dimethoxybut-3-en-2-one (46)
4-(Dimethoxymethyl)pyrimidin-2-amine (47)
5-Bromo-4-(dimethoxymethyl)pyrimidin-2-amine (48)
tert-Butyl N-[5-Bromo-4-(dimethoxymethyl)pyrimidin-2-yl]-N-[(tert-butoxy)carbonyl]carbamate (49)
(E)-N,N-Dimethyl-N′-(5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-4-(trifluoromethyl)pyridin-2-yl)formimidamide (50)
(S)-4-(2-Chloro-5,5a,6,7,8,9-hexahydropyrimido[5,4-b]indolizin-4-yl)morpholine (51a)
(R)-4-(2-Chloro-5,5a,6,7,8,9-hexahydropyrimido[5,4-b]indolizin-4-yl)morpholine (51b)
(S)-2-Chloro-4-(piperidin-1-yl)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (52a)
(R)-2-Chloro-4-(piperidin-1-yl)-5a,6,8,9-tetrahydro-5H-pyrimido[5′,4′:4,5]pyrrolo[2,1-c][1,4]oxazine (52b)
(S)-Hexahydro-[1,2,3]oxathiazolo[3,4-a]pyridine 1,1-Dioxide (53a)
(R)-Hexahydro-[1,2,3]oxathiazolo[3,4-a]pyridine 1,1-Dioxide (53b)
2,4-Dichloro-6-(piperidin-1-yl)pyrimidine (54)
Structure Modeling of PI3K and mTOR Kinase Complexes
Determination of Inhibitor Dissociation Constants
Kinome Profiling

Cellular PI3K and mTOR Signaling
Pharmacokinetic Studies
Hepatocyte Stability Assay
CYP Reactive Phenotyping with Human Recombinant CYP1A1 and CYP1A2 Isoenzymes
Metabolite Identification of Compound 11b
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.9b00972.
Syntheses of bromo derivative 44 (Scheme S1), bromo derivative 49 (Scheme S2), and compounds 3a–5a and 3b–5b (Scheme S3); plasma concentration of 7b after a single po dose of 5 mg/kg in rats (Table S1); brain concentration of 7b after a single po dose of 5 mg/kg in rats (Table S2); plasma concentration of 12b after a single po dose of 5 mg/kg in rats (Table S3); brain concentration of 12b after a single po dose of 5 mg/kg in rats (Table S4); stability of compound 11b (5 μM) in primary mouse, rat, dog, and human hepatocytes (Table S5); CYP1A1 and CYP1A2 metabolites identification of 11b (Table S6); proposed metabolic pathway for CYP-dependent metabolism of 11b (Figure S1); chromatogram of compound 11b incubated with CYP1A1 (60 min) (Figure S2); chromatogram of compound 11b incubated with CYP1A1 (0 min) (Figure S3); chromatogram of compound 11b incubated with CYP1A2 (60 min) (Figure S4); chromatogram of compound 11b incubated with CYP1A2 (0 min) (Figure S5); activity data and standard errors of final compounds (Table S7); activity data and standard errors of compounds for modeling (Table S8); TREEspot data visualization of KINOMEScan interactions of compound 12b, PQR620, and INK128 (Figure S6); selectivity profile calculated from KinomeScan data (Table S9); kinase interactions (KINOMEscan data) (Table S10); 1H NMR, 13C{1H} NMR, and NSI-HRMS spectra; HPLC chromatograms; chemical structures of final compounds and intermediates (PDF)
Compound 3a-PI3Kγ (PDB)
Compound 2a-mTOR (PDB)
Compound 2b-mTOR (PDB)
Compound 2a-PI3Kα (PDB)
Compound 2b-PI3Kα (PDB)
Molecular formula strings and some data (CSV)
PDB code 5JHB was used for docking of compound 3a into PI3Kγ. PDB code 4JT6 was used for docking of compounds 2a and 2b into mTOR kinase. PDB code 3ZIM was used for docking of compounds 2a and 2b into PI3Kα.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank A. Pfaltz, J. Füglistaler, C. Meyer, J. Schwarte, and E. Teillet for advice, discussions, and contributions to synthetic efforts, and we thank S. Bünger for technical assistance. This work was supported by the Swiss Commission for Technology and Innovation (CTI) by PFLS-LS Grants 14032.1, 15811.2, and 17241.1; the Stiftung für Krebsbekämpfung Grant 341; and Swiss National Science Foundation Grants 310030_153211 and 316030_133860 (to M.P.W.).
mTOR | mechanistical (or mammalian) target of rapamycin |
TORC1 | mTOR complex 1 |
TORC2 | mTOR complex 2 |
PI3K | phosphoinositide 3-kinase |
PKB | protein kinase B/Akt |
S6RP | ribosomal protein S6 |
S6K | p70 S6 kinase |
VPS34 | vacuolar protein sorting 34 (the class III PI3K) |
TORKi | mTOR kinase inhibitor |
PK | pharmacokinetic |
TR-FRET | time-resolved Förster resonance energy transfer |
References
This article references 49 other publications.
- 1Wymann, M. P.; Schneiter, R. Lipid signalling in disease. Nat. Rev. Mol. Cell Biol. 2008, 9 (2), 162– 176, DOI: 10.1038/nrm2335Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOgug%253D%253D&md5=c96aabfdfa8e93c9e98386f1a407de85Lipid signalling in diseaseWymann, Matthias P.; Schneiter, RogerNature Reviews Molecular Cell Biology (2008), 9 (2), 162-176CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Signaling lipids such as eicosanoids, phosphoinositides, sphingolipids and fatty acids control important cellular processes, including cell proliferation, apoptosis, metab. and migration. Extracellular signals from cytokines, growth factors and nutrients control the activity of a key set of lipid-modifying enzymes: phospholipases, prostaglandin synthase, 5-lipoxygenase, phosphoinositide 3-kinase, sphingosine kinase and sphingomyelinase. These enzymes and their downstream targets constitute a complex lipid signaling network with multiple nodes of interaction and cross-regulation. Imbalances in this network contribute to the pathogenesis of human disease. Although the function of a particular signaling lipid is traditionally studied in isolation, this review attempts a more integrated overview of the key role of these signaling lipids in inflammation, cancer and metabolic disease, and discusses emerging strategies for therapeutic intervention.
- 2Yang, H.; Rudge, D. G.; Koos, J. D.; Vaidialingam, B.; Yang, H. J.; Pavletich, N. P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497 (7448), 217– 223, DOI: 10.1038/nature12122Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvFyns7k%253D&md5=07809555fbdf2004819a7f2da5727bafmTOR kinase structure, mechanism and regulationYang, Haijuan; Rudge, Derek G.; Koos, Joseph D.; Vaidialingam, Bhamini; Yang, Hyo J.; Pavletich, Nikola P.Nature (London, United Kingdom) (2013), 497 (7448), 217-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mammalian target of rapamycin (mTOR), a phosphoinositide 3-kinase-related protein kinase, controls cell growth in response to nutrients and growth factors and is frequently deregulated in cancer. Here we report co-crystal structures of a complex of truncated mTOR and mammalian lethal with SEC13 protein 8 (mLST8) with an ATP transition state mimic (MgF3-) and with ATP-site inhibitors (Torin2, PP242, and PI-103). The structures reveal an intrinsically active kinase conformation, with catalytic residues and a catalytic mechanism remarkably similar to canonical protein kinases. The active site is highly recessed owing to the FKBP12-rapamycin-binding (FRB) domain and an inhibitory helix protruding from the catalytic cleft. MTOR-activating mutations map to the structural framework that holds these elements in place, indicating that the kinase is controlled by restricted access. In vitro biochem. shows that the FRB domain acts as a gatekeeper, with its rapamycin-binding site interacting with substrates to grant them access to the restricted active site. Rapamycin-FKBP12 inhibits the kinase by directly blocking substrate recruitment and by further restricting active-site access. The structures also reveal active-site residues and conformational changes that underlie inhibitor potency and specificity.
- 3Sarbassov, D. D.; Guertin, D. A.; Ali, S. M.; Sabatini, D. M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307 (5712), 1098– 1101, DOI: 10.1126/science.1106148Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlSrtbY%253D&md5=de758fd8128561e34301e67a42ae13a6Phosphorylation and Regulation of Akt/PKB by the Rictor-mTOR ComplexSarbassov, Dos D.; Guertin, David A.; Ali, Siraj M.; Sabatini, David M.Science (Washington, DC, United States) (2005), 307 (5712), 1098-1101CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Deregulation of Akt/protein kinase B (PKB) is implicated in the pathogenesis of cancer and diabetes. Akt/PKB activation requires the phosphorylation of Thr308 in the activation loop by the phosphoinositide-dependent kinase 1 (PDK1) and Ser473 within the carboxyl-terminal hydrophobic motif by an unknown kinase. We show that in Drosophila and human cells the target of rapamycin (TOR) kinase and its assocd. protein rictor are necessary for Ser473 phosphorylation and that a redn. in rictor or mammalian TOR (mTOR) expression inhibited an Akt/PKB effector. The rictor-mTOR complex directly phosphorylated Akt/PKB on Ser473 in vitro and facilitated Thr308 phosphorylation by PDK1. Rictor-mTOR may serve as a drug target in tumors that have lost the expression of PTEN, a tumor suppressor that opposes Akt/PKB activation.
- 4Feng, J.; Park, J.; Cron, P.; Hess, D.; Hemmings, B. A. Identification of a PKB/Akt hydrophobic motif Ser-473 kinase as DNA-dependent protein kinase. J. Biol. Chem. 2004, 279 (39), 41189– 41196, DOI: 10.1074/jbc.M406731200Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXnslWms70%253D&md5=b6bad6828d91d47e2b36f537ca6d5f85Identification of a PKB/Akt Hydrophobic Motif Ser-473 Kinase as DNA-dependent Protein KinaseFeng, Jianhua; Park, Jongsun; Cron, Peter; Hess, Daniel; Hemmings, Brian A.Journal of Biological Chemistry (2004), 279 (39), 41189-41196CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Full activation of protein kinase B (PKB)/Akt requires phosphorylation on Thr-308 and Ser-473 by 3-phosphoinositide-dependent kinase-1 (PDK1) and Ser-473 kinase (S473K), resp. Although PDK1 has been well characterized, the identification of the S473K remains controversial. A major PKB Ser-473 kinase activity was purified from the membrane fraction of HEK293 cells and found to be DNA-dependent protein kinase (DNA-PK). DNA-PK co-localized and assocd. with PKB at the plasma membrane. In vitro, DNA-PK phosphorylated PKB on Ser-473, resulting in a ∼10-fold enhancement of PKB activity. Knockdown of DNA-PK by small interfering RNA inhibited Ser-473 phosphorylation induced by insulin and pervanadate. DNA-PK-deficient glioblastoma cells did not respond to insulin at the level of Ser-473 phosphorylation; this effect was restored by complementation with the human PRKDC gene. We conclude that DNA-PK is a long sought after kinase responsible for the Ser-473 phosphorylation step in the activation of PKB.
- 5Wymann, M. P.; Marone, R. Phosphoinositide 3-kinase in disease: timing, location, and scaffolding. Curr. Opin. Cell Biol. 2005, 17 (2), 141– 149, DOI: 10.1016/j.ceb.2005.02.011Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVCqtb4%253D&md5=d126dfae131b86e73e8842ec9149fd01Phosphoinositide 3-kinase in disease: timing, location, and scaffoldingWymann, Matthias P.; Marone, RominaCurrent Opinion in Cell Biology (2005), 17 (2), 141-149CODEN: COCBE3; ISSN:0955-0674. (Elsevier Ltd.)A review. When PI3Ks are deregulated by aberrant surface receptors or modulators, accumulation of PtdIns(3,4,5)P3 leads to increased cell growth, proliferation and contact-independent survival. The PI3K/PKB/TOR axis controls protein synthesis and growth, while PtdIns(3,4,5)P3-mediated activation of Rho GTPases directs cell motility. PI3K activity has been linked to the formation of tumors, metastasis, chronic inflammation, allergy and cardiovascular disease. Although increased PtdIns(3,4,5)P3 is a well-established cause of disease, it is seldom known which PI3K isoform is implied. Recent work has demonstrated that PI3Kγ contributes to the control of cAMP levels in the cardiac system, where the protein acts as a scaffold, but not as a lipid kinase.
- 6Bozulic, L.; Hemmings, B. A. PIKKing on PKB: regulation of PKB activity by phosphorylation. Curr. Opin. Cell Biol. 2009, 21 (2), 256– 261, DOI: 10.1016/j.ceb.2009.02.002Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXksVClu7Y%253D&md5=0b7a3e6ea561f8479701a01133b5d30cPIKKing on PKB: regulation of PKB activity by phosphorylationBozulic, Lana; Hemmings, Brian A.Current Opinion in Cell Biology (2009), 21 (2), 256-261CODEN: COCBE3; ISSN:0955-0674. (Elsevier B.V.)A review. Protein kinase B (PKB)/Akt kinase is a key regulator of a wide range of cellular processes including growth, proliferation and survival. PKB is clearly a crucial signaling mol. and extensive research efforts aim to understand its regulation and action. Recent studies of the regulation of PKB activity by hydrophobic motif phosphorylation have yielded several exciting findings about members of the phosphatidylinositol 3-kinase (PI3K)-like family of kinases (PIKKs) acting as PKB regulators. Mammalian target of rapamycin complex 2 (mTORC2) and DNA-dependent protein kinase (DNA-PK) can both phosphorylate Ser-473 and activate PKB. This review concerns PKB regulation by mTORC2 and DNA-PK in a stimulus-dependent and context-dependent manner and the possible implications of this for PKB activity, substrate specificity, and therapeutic intervention.
- 7Magnuson, B.; Ekim, B.; Fingar, D. C. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem. J. 2012, 441 (1), 1– 21, DOI: 10.1042/BJ20110892Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1Cgs7jK&md5=5d69e3be00deef02d3fce1ed2716873dRegulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networksMagnuson, Brian; Ekim, Bilgen; Fingar, Diane C.Biochemical Journal (2012), 441 (1), 1-21CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)A review. The ribosomal protein S6K (S6 kinase) represents an extensively studied effector of the TORC1 [TOR (target of rapamycin) complex 1], which possesses important yet incompletely defined roles in cellular and organismal physiol. TORC1 functions as an environmental sensor by integrating signals derived from diverse environmental cues to promote anabolic and inhibit catabolic cellular functions. mTORC1 (mammalian TORC1) phosphorylates and activates S6K1 and S6K2, whose first identified substrate was rpS6 (ribosomal protein S6), a component of the 40S ribosome. Studies over the past decade have uncovered a no. of addnl. S6K1 substrates, revealing multiple levels at which the mTORC1-S6K1 axis regulates cell physiol. The results thus far indicate that the mTORC1-S6K1 axis controls fundamental cellular processes, including transcription, translation, protein and lipid synthesis, cell growth/size and cell metab. In the present review we summarize the regulation of S6Ks, their cellular substrates and functions, and their integration within rapidly expanding mTOR (mammalian TOR) signalling networks. Although our understanding of the role of mTORC1-S6K1 signalling in physiol. remains in its infancy, evidence indicates that this signalling axis controls, at least in part, glucose homeostasis, insulin sensitivity, adipocyte metab., body mass and energy balance, tissue and organ size, learning, memory and aging. As dysregulation of this signalling axis contributes to diverse disease states, improved understanding of S6K regulation and function within mTOR signalling networks may enable the development of novel therapeutics.
- 8Laplante, M.; Sabatini, D. M. mTOR signaling in growth control and disease. Cell 2012, 149 (2), 274– 293, DOI: 10.1016/j.cell.2012.03.017Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xls1eguro%253D&md5=d1adac8ec64da63358e0af26a17ceb4emTOR signaling in growth control and diseaseLaplante, Mathieu; Sabatini, David M.Cell (Cambridge, MA, United States) (2012), 149 (2), 274-293CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The mechanistic target of rapamycin (mTOR) signaling pathway senses and integrates a variety of environmental cues to regulate organismal growth and homeostasis. The pathway regulates many major cellular processes and is implicated in an increasing no. of pathol. conditions, including cancer, obesity, type 2 diabetes, and neurodegeneration. Here, we review recent advances in our understanding of the mTOR pathway and its role in health, disease, and aging. We further discuss pharmacol. approaches to treat human pathologies linked to mTOR deregulation.
- 9Saxton, R. A.; Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 2017, 168 (6), 960– 976, DOI: 10.1016/j.cell.2017.02.004Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkt1Ogtb4%253D&md5=3fdee3d04bf88d3aafc532d4e2f1e2dcmTOR signaling in growth, metabolism, and diseaseSaxton, Robert A.; Sabatini, David M.Cell (Cambridge, MA, United States) (2017), 168 (6), 960-976CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The mechanistic target of rapamycin (mTOR) coordinates eukaryotic cell growth and metab. with environmental inputs, including nutrients and growth factors. Extensive research over the past two decades has established a central role for mTOR in regulating many fundamental cell processes, from protein synthesis to autophagy, and deregulated mTOR signaling is implicated in the progression of cancer and diabetes, as well as the aging process. Here, we review recent advances in our understanding of mTOR function, regulation, and importance in mammalian physiol. We also highlight how the mTOR signaling network contributes to human disease and discuss the current and future prospects for therapeutically targeting mTOR in the clinic.
- 10Vivanco, I.; Sawyers, C. L. The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat. Rev. Cancer 2002, 2 (7), 489– 501, DOI: 10.1038/nrc839Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XkvFKltLs%253D&md5=c6762d32f9f1281632f4ccdbcd29268cThe phosphatidylinositol 3-Kinase-AKT pathway in human cancerVivanco, Igor; Sawyers, Charles L.Nature Reviews Cancer (2002), 2 (7), 489-501CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. One signal that is overactivated in a wide range of tumor types is the prodn. of a phospholipid, phosphatidylinositol (3,4,5) trisphosphate, by phosphatidylinositol 3-kinase (PI3K). This lipid and the protein kinase that is activated by it, AKT, trigger a cascade of responses, from cell growth and proliferation to survival and motility, that drive tumor progression. Small-mol. therapeutics that block PI3K signaling might deal a severe blow to cancer cells by blocking many aspects of the tumor-cell phenotype.
- 11Marone, R.; Cmiljanovic, V.; Giese, B.; Wymann, M. P. Targeting phosphoinositide 3-kinase: moving towards therapy. Biochim. Biophys. Acta, Proteins Proteomics 2008, 1784 (1), 159– 185, DOI: 10.1016/j.bbapap.2007.10.003Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXptlerug%253D%253D&md5=bf9f259b3da61067631823656658e0f7Targeting phosphoinositide 3-kinase-Moving towards therapyMarone, Romina; Cmiljanovic, Vladimir; Giese, Bernd; Wymann, Matthias P.Biochimica et Biophysica Acta, Proteins and Proteomics (2008), 1784 (1), 159-185CODEN: BBAPBW; ISSN:1570-9639. (Elsevier Ltd.)A review. Phosphoinositide 3-kinases (PI3K) orchestrate cell responses including mitogenic signaling, cell survival and growth, metabolic control, vesicular trafficking, degranulation, cytoskeletal rearrangement and migration. Deregulation of the PI3K pathway occurs by activating mutations in growth factor receptors or the PIK3CA locus coding for PI3Kα, by loss of function of the lipid phosphatase and tensin homolog deleted in chromosome ten (PTEN/MMAC/TEP1), by the up-regulation of protein kinase B (PKB/Akt), or the impairment of the tuberous sclerosis complex (TSC1/2). All these events are linked to growth and proliferation, and have thus prompted a significant interest in the pharmaceutical targeting of the PI3K pathway in cancer. Genetic targeting of PI3Kγ (p110γ) and PI3Kδ (p110δ) in mice has underlined a central role of these PI3K isoforms in inflammation and allergy, as they modulate chemotaxis of leukocytes and degranulation in mast cells. Proof-of-concept mols. selective for PI3Kγ have already successfully alleviated disease progress in murine models of rheumatoid arthritis and lupus erythematosus. As targeting PI3K moves forward to therapy of chronic, non-fatal disease, safety concerns for PI3K inhibitors increase. Many of the present inhibitor series interfere with target of rapamycin (TOR), DNA-dependent protein kinase (DNA-PKcs) and activity of the ataxia telangiectasia mutated gene product (ATM). Here we review the current disease-relevant knowledge for isoform-specific PI3K function in the above mentioned diseases, and review the progress of > 400 recent patents covering pharmaceutical targeting of PI3K. Currently, several drugs targeting the PI3K pathway have entered clin. trials (phase I) for solid tumors and suppression of tissue damage after myocardial infarction (phases I,II).
- 12Wymann, M. PI3Ks—Drug Targets in Inflammation and Cancer. In Phosphoinositides I: Enzymes of Synthesis and Degradation; Balla, T., Wymann, M., York, J. D., Eds.; Springer: Dordrecht, The Netherlands, 2012; pp 111– 181.Google ScholarThere is no corresponding record for this reference.
- 13Choi, J.; Chen, J.; Schreiber, S. L.; Clardy, J. Structure of the FKBP1 2-rapamycin complex interacting with the binding domain of human FRAP. Science 1996, 273 (5272), 239– 242, DOI: 10.1126/science.273.5272.239Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xkt1Crs7k%253D&md5=c72a82e14c09d9a705215b4ac0525336Structure of the FKBP12-rapamycin complex interacting with the binding domain of human FRAPChoi, Jungwon; Chen, Jie; Schreiber, Stuart L.; Clardy, JonScience (Washington, D. C.) (1996), 273 (5272), 239-242CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Rapamycin, a potent immunosuppressive agent, binds two proteins: the FK506-binding protein (FKBP12) and the FKBP-rapamycin-assocd. protein (FRAP). A crystal structure of the ternary complex of human FKBP12, rapamycin, and the FKBP12-rapamycin-binding (FRB) domain of human FRAP at a resoln. of 2.7 angstroms revealed the two proteins bound together as a result of the ability of rapamycin to occupy two different hydrophobic binding pockets simultaneously. The structure shows extensive interactions between rapamycin and both proteins, but fewer interactions between the proteins. The structure of the FRB domain of FRAP clarifies both rapamycin-independent and -dependent effects obsd. for mutants of FRAP and its homologs in the family of proteins related to the ataxia-telangiectasis mutant gene product, and it illustrates how a small cell-permeable mol. can mediate protein dimerization.
- 14Hudes, G.; Carducci, M.; Tomczak, P.; Dutcher, J.; Figlin, R.; Kapoor, A.; Staroslawska, E.; Sosman, J.; McDermott, D.; Bodrogi, I.; Kovacevic, Z.; Lesovoy, V.; Schmidt-Wolf, I. G. H.; Barbarash, O.; Gokmen, E.; O’Toole, T.; Lustgarten, S.; Moore, L.; Motzer, R. J. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 2007, 356 (22), 2271– 2281, DOI: 10.1056/NEJMoa066838Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtVKkurs%253D&md5=2fc7685df067fd6ea813483610e89dc8Temsirolimus, interferon alfa, or both for advanced renal-cell carcinomaHudes, Gary; Carducci, Michael; Tomczak, Piotr; Dutcher, Janice; Figlin, Robert; Kapoor, Anil; Staroslawska, Elzbieta; Sosman, Jeffrey; McDermott, David; Bodrogi, Istvan; Kovacevic, Zoran; Lesovoy, Vladimir; Schmidt-Wolf, Ingo G. H.; Barbarash, Olga; Gokmen, Erhan; O'Toole, Timothy; Lustgarten, Stephanie; Moore, Laurence; Motzer, Robert J.New England Journal of Medicine (2007), 356 (22), 2271-2281CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)Interferon alfa is widely used for metastatic renal-cell carcinoma but has limited efficacy and tolerability. Temsirolimus, a specific inhibitor of the mammalian target of rapamycin kinase, may benefit patients with this disease. In this multicenter, phase 3 trial, we randomly assigned 626 patients with previously untreated, poor-prognosis metastatic renal-cell carcinoma to receive 25 mg of i.v. temsirolimus weekly, 3 million U of interferon alfa (with an increase to 18 million U) s.c. three times weekly, or combination therapy with 15 mg of temsirolimus weekly plus 6 million U of interferon alfa three times weekly. The primary end point was overall survival in comparisons of the temsirolimus group and the combination-therapy group with the interferon group. RESULTS Patients who received temsirolimus alone had longer overall survival (hazard ratio for death, 0.73; 95% confidence interval [CI], 0.58 to 0.92; P = 0.008) and progression-free survival (P < 0.001) than did patients who received interferon alone. Overall survival in the combination-therapy group did not differ significantly from that in the interferon group (hazard ratio, 0.96; 95% CI, 0.76 to 1.20; P = 0.70). Median overall survival times in the interferon group, the temsirolimus group, and the combination-therapy group were 7.3, 10.9, and 8.4 mo, resp. Rash, peripheral edema, hyperglycemia, and hyperlipidemia were more common in the temsirolimus group, whereas asthenia was more common in the interferon group. There were fewer patients with serious adverse events in the temsirolimus group than in the interferon group (P = 0.02). As compared with interferon alfa, temsirolimus improved overall survival among patients with metastatic renal-cell carcinoma and a poor prognosis. The addn. of temsirolimus to interferon did not improve survival. (ClinicalTrials.gov no., NCT00065468.).
- 15Motzer, R. J.; Escudier, B.; Oudard, S.; Hutson, T. E.; Porta, C.; Bracarda, S.; Grünwald, V.; Thompson, J. A.; Figlin, R. A.; Hollaender, N.; Urbanowitz, G.; Berg, W. J.; Kay, A.; Lebwohl, D.; Ravaud, A. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008, 372 (9637), 449– 456, DOI: 10.1016/S0140-6736(08)61039-9Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1GmsLY%253D&md5=cbfa5d207cfab693b3bccd24963380f0Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomized, placebo-controlled phase III trialMotzer, Robert J.; Escudier, Bernard; Oudard, Stephane; Hutson, Thomas E.; Porta, Camillo; Bracarda, Sergio; Gruenwald, Viktor; Thompson, John A.; Figlin, Robert A.; Hollaender, Norbert; Urbanowitz, Gladys; Berg, William J.; Kay, Andrea; Lebwohl, David; Ravaud, AlainLancet (2008), 372 (9637), 449-456CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Everolimus (RAD001) is an orally administered inhibitor of the mammalian target of rapamycin (mTOR), a therapeutic target for metastatic renal cell carcinoma. We did a phase III, randomized, double-blind, placebo-controlled trial of everolimus in patients with metastatic renal cell carcinoma whose disease had progressed on vascular endothelial growth factor-targeted therapy. Patients with metastatic renal cell carcinoma which had progressed on sunitinib, sorafenib, or both, were randomly assigned in a two to one ratio to receive everolimus 10 mg once daily (n=272) or placebo (n=138), in conjunction with best supportive care. Randomisation was done centrally via an interactive voice response system using a validated computer system, and was stratified by Memorial Sloan-Kettering Cancer Center prognostic score and previous anticancer therapy, with a permuted block size of six. The primary endpoint was progression-free survival, assessed via a blinded, independent central review. The study was designed to be terminated after 290 events of progression. Anal. was by intention to treat. This study is registered with, no. All randomized patients were included in efficacy analyses. The results of the second interim anal. indicated a significant difference in efficacy between arms and the trial was thus halted early after 191 progression events had been obsd. (101 [37%] events in the everolimus group, 90 [65%] in the placebo group; hazard ratio 0.30, 95% CI 0.22-0.40, p<0.0001; median progression-free survival 4.0 [95% CI 3.7-5.5] vs 1.9 [1.8-1.9] months). Stomatitis (107 [40%] patients in the everolimus group vs 11 [8%] in the placebo group), rash (66 [25%] vs six [4%]), and fatigue (53 [20%] vs 22 [16%]) were the most commonly reported adverse events, but were mostly mild or moderate in severity. Pneumonitis (any grade) was detected in 22 (8%) patients in the everolimus group, of whom eight had pneumonitis of grade 3 severity. Treatment with everolimus prolongs progression-free survival relative to placebo in patients with metastatic renal cell carcinoma that had progressed on other targeted therapies. Novartis Oncol.
- 16Jerusalem, G.; Rorive, A.; Collignon, J. Use of mTOR inhibitors in the treatment of breast cancer: an evaluation of factors that influence patient outcomes. Breast Cancer: Targets Ther. 2014, 6, 43– 57, DOI: 10.2147/BCTT.S38679Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs12gsrjE&md5=9a12c70c7c917baa41d282bc135b12afUse of mTOR inhibitors in the treatment of breast cancer: an evaluation of factors that influence patient outcomesJerusalem, Guy; Rorive, Andree; Collignon, JoelleBreast Cancer: Targets and Therapy (2014), 6 (), 43-57, 15CODEN: BCTTA9; ISSN:1179-1314. (Dove Medical Press Ltd.)A review. Many systemic treatment options are available for advanced breast cancer, including endocrine therapy, chemotherapy, anti-human epidermal growth factor receptor 2 (HER2) therapy, and other targeted agents. Recently, everolimus, a mammalian target of rapamycin (mTOR) inhibitor, combined with exemestane, an aromatase inhibitor, has been approved in Europe and the USA for patients suffering from estrogen receptor-pos., HER2-neg. advanced breast cancer previously treated by a nonsteroidal aromatase inhibitor, based on the results of BOLERO-2 (Breast cancer trials of OraL EveROlimus). This study showed a statistically significant and clin. meaningful improvement in median progression-free survival. Results concerning the impact on overall survival are expected in the near future. This clin. oriented review focuses on the use of mTOR inhibitors in breast cancer. Results reported with first-generation mTOR inhibitors (ridaforolimus, temsirolimus, everolimus) are discussed. The current and potential role of mTOR inhibitors is reported according to breast cancer subtype (estrogen receptor-pos. HER2-neg., triple-neg., and HER2-pos. ER-pos./neg. disease). Everolimus is currently being evaluated in the adjuvant setting in high-risk estrogen receptor-pos., HER2-neg. early breast cancer. Continuing mTOR inhibition or alternatively administering other drugs targeting the phosphatidylinositol-3-kinase/protein kinase B-mTOR pathway after progression on treatments including an mTOR inhibitor is under evaluation. Potential biomarkers to select patients showing a more pronounced benefit are reviewed, but we are not currently using these biomarkers in routine practice. Subgroup anal. of BOLERO 2 has shown that the benefit is consistent in all subgroups and that it is impossible to select patients not benefiting from addn. of everolimus to exemestane. Side effects and impact on quality of life are other important issues discussed in this review. Second-generation mTOR inhibitors and dual mTOR-phosphatidylinositol-3-kinase inhibitors are currently being evaluated in clin. trials.
- 17André, F.; O’Regan, R.; Ozguroglu, M.; Toi, M.; Xu, B.; Jerusalem, G.; Masuda, N.; Wilks, S.; Arena, F.; Isaacs, C.; Yap, Y.-S.; Papai, Z.; Lang, I.; Armstrong, A.; Lerzo, G.; White, M.; Shen, K.; Litton, J.; Chen, D.; Zhang, Y.; Ali, S.; Taran, T.; Gianni, L. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014, 15 (6), 580– 591, DOI: 10.1016/S1470-2045(14)70138-XGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlWnsr4%253D&md5=cfbb16368227cc80463ef26c2593b69dEverolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trialAndre, Fabrice; O'Regan, Ruth; Ozguroglu, Mustafa; Toi, Masakazu; Xu, Binghe; Jerusalem, Guy; Masuda, Norikazu; Wilks, Sharon; Arena, Francis; Isaacs, Claudine; Yap, Yoon-Sim; Papai, Zsuzsanna; Lang, Istvan; Armstrong, Anne; Lerzo, Guillermo; White, Michelle; Shen, Kunwei; Litton, Jennifer; Chen, David; Zhang, Yufen; Ali, Shyanne; Taran, Tetiana; Gianni, LucaLancet Oncology (2014), 15 (6), 580-591CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Disease progression in patients with HER2-pos. breast cancer receiving trastuzumab might be assocd. with activation of the PI3K/Akt/mTOR intracellular signalling pathway. We aimed to assess whether the addn. of the mTOR inhibitor everolimus to trastuzumab might restore sensitivity to trastuzumab. In this randomised, double-blind, placebo-controlled, phase 3 trial, we recruited women with HER2-pos., trastuzumab-resistant, advanced breast carcinoma who had previously received taxane therapy. Eligible patients were randomly assigned (1:1) using a central patient screening and randomization system to daily everolimus (5 mg/day) plus weekly trastuzumab (2 mg/kg) and vinorelbine (25 mg/m2) or to placebo plus trastuzumab plus vinorelbine, in 3-wk cycles, stratified by previous lapatinib use. The primary endpoint was progression-free survival (PFS) by local assessment in the intention-to-treat population. We report the final anal. for PFS; overall survival follow-up is still in progress. This trial is registered with ClinicalTrials.gov, no. NCT01007942.Between Oct 26, 2009, and May 23, 2012, 569 patients were randomly assigned to everolimus (n=284) or placebo (n=285). Median follow-up at the time of anal. was 20·2 mo (IQR 15·0-27·1). Median PFS was 7·00 mo (95% CI 6·74-8·18) with everolimus and 5·78 mo (5·49-6·90) with placebo (hazard ratio 0·78 [95% CI 0·65-0·95]; p=0·0067). The most common grade 3-4 adverse events were neutropenia (204 [73%] of 280 patients in the everolimus group vs 175 [62%] of 282 patients in the placebo group), leucopenia (106 [38%] vs 82 [29%]), anemia (53 [19%] vs 17 [6%]), febrile neutropenia (44 [16%] vs ten [4%]), stomatitis (37 [13%] vs four [1%]), and fatigue (34 [12%] vs 11 [4%]). Serious adverse events were reported in 117 (42%) patients in the everolimus group and 55 (20%) in the placebo group; two on-treatment deaths due to adverse events occurred in each group. The addn. of everolimus to trastuzumab plus vinorelbine significantly prolongs PFS in patients with trastuzumab-resistant and taxane-pretreated, HER2-pos., advanced breast cancer. The clin. benefit should be considered in the context of the adverse event profile in this population. Novartis Pharmaceuticals Corporation.
- 18Peterson, M. E. Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trial. Supportive care in cancer: official journal of the Multinational Association of Supportive Care in Cancer 2013, 21 (8), 2341– 2349, DOI: 10.1007/s00520-013-1826-3Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3snkvVKksw%253D%253D&md5=21bfc1094be58365540d6fe494138198Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trialPeterson Mary ESupportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer (2013), 21 (8), 2341-9 ISSN:.Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved for the treatment of advanced renal cell carcinoma, pancreatic neuroendocrine tumors, subependymal giant cell astrocytoma associated with tuberous sclerosis complex, renal angiomyolipoma and tuberous sclerosis complex, and, in combination with exemestane, for hormone receptor-positive HER2-negative advanced breast cancer after failure of treatment with letrozole or anastrozole. Results from the phase III BOLERO-2 trial demonstrated that everolimus in combination with exemestane provided significant clinical benefit to patients with advanced hormone receptor-positive breast cancer. Although everolimus is generally well tolerated, as with most therapies administered in an advanced cancer setting, drug-related adverse events (AEs) inevitably occur. Most common AEs observed in the everolimus studies include stomatitis, rash, infection, noninfectious pneumonitis, and hyperglycemia. Clinical awareness and early identification of such AEs by oncology nurses are essential to dosing (interruptions, reduction, and treatment discontinuation); quality of life; and, ultimately, patient outcomes. Because everolimus has already been shown to significantly improve clinical efficacy in patients with advanced breast cancer, a proactive approach to the practical management of AEs associated with this mTOR inhibitor as well as other most common AEs observed in this patient population has been reviewed and outlined here.
- 19Santulli, G.; Totary-Jain, H. Tailoring mTOR-based therapy: molecular evidence and clinical challenges. Pharmacogenomics 2013, 14 (12), 1517– 1526, DOI: 10.2217/pgs.13.143Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVWrurrN&md5=0a3798fb659843d231910151b2f148edTailoring mTOR-based therapy: molecular evidence and clinical challengesSantulli, Gaetano; Totary-Jain, HanaPharmacogenomics (2013), 14 (12), 1517-1526CODEN: PARMFL; ISSN:1462-2416. (Future Medicine Ltd.)A review. The mTOR signaling pathway integrates inputs from a variety of upstream stimuli to regulate diverse cellular processes including proliferation, growth, survival, motility, autophagy, protein synthesis and metab. The mTOR pathway is dysregulated in a no. of human pathologies including cancer, diabetes, obesity, autoimmune disorders, neurol. disease and aging. Ongoing clin. trials testing mTOR-targeted treatments no. in the hundreds and underscore its therapeutic potential. To date mTOR inhibitors are clin. approved to prevent organ rejection, to inhibit restenosis after angioplasty, and to treat several advanced cancers. In this review we discuss the continuously evolving field of mTOR pharmacogenomics, as well as highlight the emerging efforts in identifying diagnostic and prognostic markers, including miRNAs, in order to assess successful therapeutic responses.
- 20Meng, L. H.; Zheng, X. F. Toward rapamycin analog (rapalog)-based precision cancer therapy. Acta Pharmacol. Sin. 2015, 36 (10), 1163– 1169, DOI: 10.1038/aps.2015.68Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1Wls7jP&md5=af1357a49b132c2264ca74577ae2ae48Toward rapamycin analog (rapalog)-based precision cancer therapyMeng, Ling-hua; Zheng, X. F. StevenActa Pharmacologica Sinica (2015), 36 (10), 1163-1169CODEN: APSCG5; ISSN:1671-4083. (Nature Publishing Group)Rapamycin and its analogs (rapalogs) are the first generation of mTOR inhibitors, which have the same mol. scaffold, but different physiochem. properties. Rapalogs are being tested in a wide spectrum of human tumors as both monotherapy and a component of combination therapy. Among them, temsirolimus and everolimus have been approved for the treatment of breast and renal cancer. However, objective response rates with rapalogs in clin. trials are modest and variable. Identification of biomarkers predicting response to rapalogs, and discovery of drug combinations with improved efficacy and tolerated toxicity are crit. to moving this class of targeted therapeutics forward. This review focuses on the aberrations in the PI3K/mTOR pathway in human tumor cells or tissues as predictive biomarkers for rapalog efficacy. Recent results of combinational therapy using rapalogs and other anticancer drugs are documented. With the rapid development of next-generation genomic sequencing and precision medicine, rapalogs will provide greater benefits to cancer patients.
- 21Feldman, M. E.; Apsel, B.; Uotila, A.; Loewith, R.; Knight, Z. A.; Ruggero, D.; Shokat, K. M. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 2009, 7 (2), e1000038, DOI: 10.1371/journal.pbio.1000038Google ScholarThere is no corresponding record for this reference.
- 22Kang, S. A.; Pacold, M. E.; Cervantes, C. L.; Lim, D.; Lou, H. J.; Ottina, K.; Gray, N. S.; Turk, B. E.; Yaffe, M. B.; Sabatini, D. M. mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science 2013, 341 (6144), 1236566, DOI: 10.1126/science.1236566Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfktVWmsw%253D%253D&md5=904daf44e853714de3fb540a359e2546mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycinKang Seong A; Pacold Michael E; Cervantes Christopher L; Lim Daniel; Lou Hua Jane; Ottina Kathleen; Gray Nathanael S; Turk Benjamin E; Yaffe Michael B; Sabatini David MScience (New York, N.Y.) (2013), 341 (6144), 1236566 ISSN:.The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) protein kinase promotes growth and is the target of rapamycin, a clinically useful drug that also prolongs life span in model organisms. A persistent mystery is why the phosphorylation of many bona fide mTORC1 substrates is resistant to rapamycin. We find that the in vitro kinase activity of mTORC1 toward peptides encompassing established phosphorylation sites varies widely and correlates strongly with the resistance of the sites to rapamycin, as well as to nutrient and growth factor starvation within cells. Slight modifications of the sites were sufficient to alter mTORC1 activity toward them in vitro and to cause concomitant changes within cells in their sensitivity to rapamycin and starvation. Thus, the intrinsic capacity of a phosphorylation site to serve as an mTORC1 substrate, a property we call substrate quality, is a major determinant of its sensitivity to modulators of the pathway. Our results reveal a mechanism through which mTORC1 effectors can respond differentially to the same signals.
- 23Jacinto, E.; Loewith, R.; Schmidt, A.; Lin, S.; Ruegg, M. A.; Hall, A.; Hall, M. N. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. Cell Biol. 2004, 6 (11), 1122– 8, DOI: 10.1038/ncb1183Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptFentL8%253D&md5=42d53ee5d9dfc24287cf05fd29f41aa8Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitiveJacinto, Estela; Loewith, Robbie; Schmidt, Anja; Lin, Shuo; Rueegg, Markus A.; Hall, Alan; Hall, Michael N.Nature Cell Biology (2004), 6 (11), 1122-1128CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)The target of rapamycin (TOR) is a highly conserved protein kinase and a central controller of cell growth. In budding yeast, TOR is found in structurally and functionally distinct protein complexes: TORC1 and TORC2. A mammalian counterpart of TORC1 (mTORC1) has been described, but it is not known whether TORC2 is conserved in mammals. Here, the authors report that a mammalian counterpart of TORC2 (mTORC2) also exists. The mTORC2 contains mTOR, mLST8 and mAVO3, but not raptor. Like yeast TORC2, mTORC2 is rapamycin insensitive and seems to function upstream of Rho GTPases to regulate the actin cytoskeleton. The mTORC2 is not upstream of the mTORC1 effector S6K. Thus, two distinct TOR complexes constitute a primordial signaling network conserved in eukaryotic evolution to control the fundamental process of cell growth.
- 24O’Reilly, K. E.; Rojo, F.; She, Q. B.; Solit, D.; Mills, G. B.; Smith, D.; Lane, H.; Hofmann, F.; Hicklin, D. J.; Ludwig, D. L.; Baselga, J.; Rosen, N. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006, 66 (3), 1500– 1508, DOI: 10.1158/0008-5472.CAN-05-2925Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsVegtg%253D%253D&md5=758cd9999dc156bf498edf32174fc2e7mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates AktO'Reilly, Kathryn E.; Rojo, Fredi; She, Qing-Bai; Solit, David; Mills, Gordon B.; Smith, Debra; Lane, Heidi; Hofmann, Francesco; Hicklin, Daniel J.; Ludwig, Dale L.; Baselga, Jose; Rosen, NealCancer Research (2006), 66 (3), 1500-1508CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Stimulation of the insulin and insulin-like growth factor I (IGF-I) receptor activates the phosphoinositide-3-kinase/Akt/mTOR pathway causing pleiotropic cellular effects including an mTOR-dependent loss in insulin receptor substrate-1 expression leading to feedback down-regulation of signaling through the pathway. In model systems, tumors exhibiting mutational activation of phosphoinositide-3-kinase/Akt kinase, a common event in cancers, are hypersensitive to mTOR inhibitors, including rapamycin. Despite the activity in model systems, in patients, mTOR inhibitors exhibit more modest antitumor activity. We now show that mTOR inhibition induces insulin receptor substrate-1 expression and abrogates feedback inhibition of the pathway, resulting in Akt activation both in cancer cell lines and in patient tumors treated with the rapamycin deriv., RAD001. IGF-I receptor inhibition prevents rapamycin-induced Akt activation and sensitizes tumor cells to inhibition of mTOR. In contrast, IGF-I reverses the antiproliferative effects of rapamycin in serum-free medium. The data suggest that feedback down-regulation of receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this feedback loop by rapamycin may attenuate its therapeutic effects, whereas combination therapy that ablates mTOR function and prevents Akt activation may have improved antitumor activity.
- 25Liu, P.; Gan, W.; Chin, Y. R.; Ogura, K.; Guo, J.; Zhang, J.; Wang, B.; Blenis, J.; Cantley, L. C.; Toker, A.; Su, B.; Wei, W. PtdIns(3,4,5)P3-dependent activation of the mTORC2 kinase complex. Cancer Discovery 2015, 5 (11), 1194– 1209, DOI: 10.1158/2159-8290.CD-15-0460Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVWqsr3E&md5=a218e7e37d78aaa532a8ff44db230447PtdIns(3,4,5)P3-dependent activation of the mtorc2 kinase complexLiu, Pengda; Gan, Wenjian; Chin, Y. Rebecca; Ogura, Kohei; Guo, Jianping; Zhang, Jinfang; Wang, Bin; Blenis, John; Cantley, Lewis C.; Toker, Alex; Su, Bing; Wei, WenyiCancer Discovery (2015), 5 (11), 1194-1209CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)MTOR serves as a central regulator of cell growth and metab. by forming two distinct complexes, mTORC1 and mTORC2. Although mechanisms of mTORC1 activation by growth factors and amino acids have been extensively studied, the upstream regulatory mechanisms leading to mTORC2 activation remain largely elusive. Here, we report that the pleckstrin homol. (PH) domain of SIN1, an essential and unique component of mTORC2, interacts with the mTOR kinase domain to suppress mTOR activity. More importantly, PtdIns(3,4,5)P3, but not other PtdInsPn species, interacts with SIN1-PH to release its inhibition on the mTOR kinase domain, thereby triggering mTORC2 activation. Mutating crit. SIN1 residues that mediate PtdIns(3,4,5)P3 interaction inactivates mTORC2, whereas mTORC2 activity is pathol. increased by patient-derived mutations in the SIN1-PH domain, promoting cell growth and tumor formation. Together, our study unravels a PI3K-dependent mechanism for mTORC2 activation, allowing mTORC2 to activate AKT in a manner that is regulated temporally and spatially by PtdIns(3,4,5)P3. Significance: The SIN1-PH domain interacts with the mTOR kinase domain to suppress mTOR activity, and PtdIns(3,4,5)P3 binds the SIN1-PH domain to release its inhibition on the mTOR kinase domain, leading to mTORC2 activation. Cancer patient-derived SIN1-PH domain mutations gain oncogenicity by loss of suppressing mTOR activity as a means to facilitate tumorigenesis. Cancer Discov; 5(11); 1194-209. ©2015 AACR. See related commentary by Yuan and Guan, p. 1127. This article is highlighted in the In This Issue feature, p.1111.
- 26Nowak, P.; Cole, D. C.; Brooijmans, N.; Bursavich, M. G.; Curran, K. J.; Ellingboe, J. W.; Gibbons, J. J.; Hollander, I.; Hu, Y.; Kaplan, J.; Malwitz, D. J.; Toral-Barza, L.; Verheijen, J. C.; Zask, A.; Zhang, W. G.; Yu, K. Discovery of potent and selective inhibitors of the mammalian target of rapamycin (mTOR) kinase. J. Med. Chem. 2009, 52 (22), 7081– 7089, DOI: 10.1021/jm9012642Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1Oku7zL&md5=5a60157598d9eb85e4fff6dc07f01cc2Discovery of Potent and Selective Inhibitors of the Mammalian Target of Rapamycin (mTOR) KinaseNowak, Pawel; Cole, Derek C.; Brooijmans, Natasja; Curran, Kevin J.; Ellingboe, John W.; Gibbons, James J.; Hollander, Irwin; Hu, Yong Bo; Kaplan, Joshua; Malwitz, David J.; Toral-Barza, Lourdes; Verheijen, Jeroen C.; Zask, Arie; Zhang, Wei-Guo; Yu, KerJournal of Medicinal Chemistry (2009), 52 (22), 7081-7089CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The mammalian target of rapamycin (mTOR) is a central regulator of cell growth, metab., and angiogenesis and an emerging target in cancer research. High throughput screening (HTS) of our compd. collection led to the identification of 3-(4-morpholin-4-yl-1-piperidin-4-yl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenol (5a), a modestly potent and nonselective inhibitor of mTOR and phosphoinositide 3-kinase (PI3K). Optimization of compd. 5a, employing an mTOR homol. model based on an X-ray crystal structure of closely related PI3Kγ led to the discovery of 6-(1H-indol-5-yl)-4-morpholin-4-yl-1-[1-(pyridin-3-ylmethyl)piperidin-4-yl]-1H-pyrazolo[3,4-d]pyrimidine (5u), a potent and selective mTOR inhibitor (mTOR IC50 = 9 nM; PI3Kα IC50 = 1962 nM). Compd. 5u selectively inhibited cellular biomarker of mTORC1 (P-S6K, P-4EBP1) and mTORC2 (P-AKT S473) over the biomarker of PI3K/PDK1 (P-AKT T308) and did not inhibit PI3K-related kinases (PIKKs) in cellular assays. These pyrazolopyrimidines represent an exciting new series of mTOR-selective inhibitors with potential for development for cancer therapy.
- 27Jin, Z.; Niu, H.; Wang, X.; Zhang, L.; Wang, Q.; Yang, A. Preclinical study of CC223 as a potential anti-ovarian cancer agent. Oncotarget 2017, 8 (35), 58469– 58479, DOI: 10.18632/oncotarget.17753Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M%252FhtlOjuw%253D%253D&md5=091fdb75cec2ddb9018ba74c124041e8Preclinical study of CC223 as a potential anti-ovarian cancer agentJin Zhenzhen; Niu Huanfu; Wang Xuenan; Wang Qin; Yang Aijun; Zhang LeiOncotarget (2017), 8 (35), 58469-58479 ISSN:.Aberrant activation of mTOR contributes to ovarian cancer progression. CC223 is a novel and potent mTOR kinase inhibitor. The current study tested its activity against human ovarian cancer cells. We showed that CC223, at nM concentrations, inhibited survival and proliferation of established/primary human ovarian cancer cells. Further, significant apoptosis activation was observed in CC223-treated ovarian cancer cells. CC223 disrupted assembly of mTOR complex 1 (mTORC1) and mTORC2 in SKOV3 cells. Meanwhile, activation of mTORC1 and mTORC2 was almost completely blocked by CC223. Intriguingly, restoring mTOR activation by introduction of a constitutively-active Akt1 only partially inhibited CC223-induced cytotoxicity in SKOV3 cells. Further studies showed that CC223 inhibited sphingosine kinase 1 (SphK1) activity and induced reactive oxygen species (ROS) production in SKOV3 cells. At last, oral administration of CC223 potently inhibited SKOV3 xenografted tumor growth in nude mice. The results of this study imply that CC223 could be further studied as a potential anti-ovarian cancer agent.
- 28Slotkin, E. K.; Patwardhan, P. P.; Vasudeva, S. D.; de Stanchina, E.; Tap, W. D.; Schwartz, G. K. MLN0128, an ATP-competitive mTOR kinase inhibitor with potent in vitro and in vivo antitumor activity, as potential therapy for bone and soft-tissue sarcoma. Mol. Cancer Ther. 2015, 14 (2), 395– 406, DOI: 10.1158/1535-7163.MCT-14-0711Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitlOjsbo%253D&md5=7c1d867aae6e5ca99c63b554295a539aMLN0128, an ATP-Competitive mTOR Kinase Inhibitor with Potent In Vitro and In Vivo Antitumor Activity, as Potential Therapy for Bone and Soft-Tissue SarcomaSlotkin, Emily K.; Patwardhan, Parag P.; Vasudeva, Shyamprasad D.; de Stanchina, Elisa; Tap, William D.; Schwartz, Gary K.Molecular Cancer Therapeutics (2015), 14 (2), 395-406CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that exists in two complexes (mTORC1 and mTORC2) and integrates extracellular and intracellular signals to act as a master regulator of cell growth, survival, and metab. The PI3K/AKT/mTOR prosurvival pathway is often dysregulated in multiple sarcoma subtypes. First-generation allosteric inhibitors of mTORC1 (rapalogues) have been extensively tested with great preclin. promise, but have had limited clin. utility. Here, we report that MLN0128, a second-generation, ATP-competitive, pan-mTOR kinase inhibitor, acts on both mTORC1 and mTORC2 and has potent in vitro and in vivo antitumor activity in multiple sarcoma subtypes. In vitro, MLN0128 inhibits mTORC1/2 targets in a concn.-dependent fashion and shows striking antiproliferative effect in rhabdomyosarcoma (RMS), Ewing sarcoma, malignant peripheral nerve sheath tumor, synovial sarcoma, osteosarcoma, and liposarcoma. Unlike rapamycin, MLN0128 inhibits phosphorylation of 4EBP1 and NDRG1 as well as prevents the reactivation of pAKT that occurs via neg. feedback release with mTORC1 inhibition alone. In xenograft models, MLN0128 treatment results in suppression of tumor growth with two dosing schedules (1 mg/kg daily and 3 mg/kg b.i.d. t.i.w.). At the 3 mg/kg dosing schedule, MLN0128 treatment results in significantly better tumor growth suppression than rapamycin in RMS and Ewing sarcoma models. In addn., MLN0128 induces apoptosis in models of RMS both in vitro and in vivo. Results from our study strongly suggest that MLN0128 treatment should be explored further as potential therapy for sarcoma. Mol Cancer Ther; 14(2); 395-406. ©2014 AACR.
- 29Pike, K. G.; Malagu, K.; Hummersone, M. G.; Menear, K. A.; Duggan, H. M.; Gomez, S.; Martin, N. M.; Ruston, L.; Pass, S. L.; Pass, M. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014. Bioorg. Med. Chem. Lett. 2013, 23 (5), 1212– 1216, DOI: 10.1016/j.bmcl.2013.01.019Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFajs7c%253D&md5=325cb45f84bb4357005315ab3b27c0aaOptimization of potent and selective dual mTORC1 and mTORC2 inhibitors: The discovery of AZD8055 and AZD2014Pike, Kurt G.; Malagu, Karine; Hummersone, Marc G.; Menear, Keith A.; Duggan, Heather M. E.; Gomez, Sylvie; Martin, Niall M. B.; Ruston, Linette; Pass, Sarah L.; Pass, MartinBioorganic & Medicinal Chemistry Letters (2013), 23 (5), 1212-1216CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The optimization of a potent and highly selective series of dual mTORC1 and mTORC2 inhibitors is described. An initial focus on improving cellular potency while maintaining or improving other key parameters, such as aq. soly. and margins over hERG IC50, led to the discovery of the clin. candidate AZD8055. Further optimization, particularly aimed at reducing the rate of metab. in human hepatocyte incubations, resulted in the discovery of the clin. candidate AZD2014.
- 30Lee, J. S.; Vo, T. T.; Fruman, D. A. Targeting mTOR for the treatment of B cell malignancies. Br. J. Clin. Pharmacol. 2016, 82 (5), 1213– 1228, DOI: 10.1111/bcp.12888Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1Gqsr7L&md5=0ae355bfde2b1b275eb2bdfd55b4cbabTargeting mTOR for the treatment of B cell malignanciesLee, Jong-Hoon Scott; Vo, Thanh-Trang; Fruman, David A.British Journal of Clinical Pharmacology (2016), 82 (5), 1213-1228CODEN: BCPHBM; ISSN:1365-2125. (Wiley-Blackwell)Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions as a key regulator of cell growth, division and survival. Many haematol. malignancies exhibit elevated or aberrant mTOR activation, supporting the launch of numerous clin. trials aimed at evaluating the potential of single agent mTOR-targeted therapies. While promising early clin. data using allosteric mTOR inhibitors (rapamycin and its derivs., rapalogs) have suggested activity in a subset of haematol. malignancies, these agents have shown limited efficacy in most contexts. Whether the efficacy of these partial mTOR inhibitors might be enhanced by more complete target inhibition is being actively addressed with second generation ATP-competitive mTOR kinase inhibitors (TOR-KIs), which have only recently entered clin. trials. However, emerging preclin. data suggest that despite their biochem. advantage over rapalogs, TOR-KIs may retain a primarily cytostatic response. Rather, combinations of mTOR inhibition with other targeted therapies have demonstrated promising efficacy in several preclin. models. This review investigates the current status of rapalogs and TOR-KIs in B cell malignancies, with an emphasis on emerging preclin. evidence of synergistic combinations involving mTOR inhibition.
- 31Rageot, D.; Bohnacker, T.; Melone, A.; Langlois, J. B.; Borsari, C.; Hillmann, P.; Sele, A. M.; Beaufils, F.; Zvelebil, M.; Hebeisen, P.; Loscher, W.; Burke, J.; Fabbro, D.; Wymann, M. P. Discovery and preclinical characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoro methyl)pyridin-2-amine (PQR620), a highly potent and selective mTORC1/2 inhibitor for cancer and neurological disorders. J. Med. Chem. 2018, 61 (22), 10084– 10105, DOI: 10.1021/acs.jmedchem.8b01262Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvF2mtLjO&md5=e6ebf8147d6ba041285eb6538ade46abDiscovery and Preclinical Characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoromethyl)pyridin-2-amine (PQR620), a Highly Potent and Selective mTORC1/2 Inhibitor for Cancer and Neurological DisordersRageot, Denise; Bohnacker, Thomas; Melone, Anna; Langlois, Jean-Baptiste; Borsari, Chiara; Hillmann, Petra; Sele, Alexander M.; Beaufils, Florent; Zvelebil, Marketa; Hebeisen, Paul; Loscher, Wolfgang; Burke, John; Fabbro, Doriano; Wymann, Matthias P.Journal of Medicinal Chemistry (2018), 61 (22), 10084-10105CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mechanistic target of rapamycin (mTOR) promotes cell proliferation, growth, and survival and is overactivated in many tumors and central nervous system disorders. I is a novel, potent, selective, and brain penetrable inhibitor of mTORC1/2 kinase. I showed excellent selectivity for mTOR over PI3K and protein kinases and efficiently prevented cancer cell growth in a 66 cancer cell line panel. In C57BL/6J and Sprague-Dawley mice, max. concn. (Cmax) in plasma and brain was reached after 30 min, with a half-life (t1/2) > 5 h. In an ovarian carcinoma mouse xenograft model (OVCAR-3), daily dosing of I inhibited tumor growth significantly. Moreover, I attenuated epileptic seizures in a tuberous sclerosis complex (TSC) mouse model. In conclusion, I inhibits mTOR kinase potently and selectively, shows antitumor effects in vitro and in vivo, and promises advantages in CNS indications due to its brain/plasma distribution ratio.
- 32Bohnacker, T.; Prota, A. E.; Beaufils, F.; Burke, J. E.; Melone, A.; Inglis, A. J.; Rageot, D.; Sele, A. M.; Cmiljanovic, V.; Cmiljanovic, N.; Bargsten, K.; Aher, A.; Akhmanova, A.; Diaz, J. F.; Fabbro, D.; Zvelebil, M.; Williams, R. L.; Steinmetz, M. O.; Wymann, M. P. Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat. Commun. 2017, 8, 14683, DOI: 10.1038/ncomms14683Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czksV2ruw%253D%253D&md5=97d27a7d83bf6ab6094a587565f82bf6Deconvolution of Buparlisib's mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic interventionBohnacker Thomas; Beaufils Florent; Melone Anna; Rageot Denise; Sele Alexander M; Cmiljanovic Vladimir; Cmiljanovic Natasa; Wymann Matthias P; Prota Andrea E; Bargsten Katja; Steinmetz Michel O; Burke John E; Inglis Alison J; Williams Roger L; Aher Amol; Akhmanova Anna; Diaz J Fernando; Fabbro Doriano; Zvelebil MarketaNature communications (2017), 8 (), 14683 ISSN:.BKM120 (Buparlisib) is one of the most advanced phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of cancer, but it interferes as an off-target effect with microtubule polymerization. Here, we developed two chemical derivatives that differ from BKM120 by only one atom. We show that these minute changes separate the dual activity of BKM120 into discrete PI3K and tubulin inhibitors. Analysis of the compounds cellular growth arrest phenotypes and microtubule dynamics suggest that the antiproliferative activity of BKM120 is mainly due to microtubule-dependent cytotoxicity rather than through inhibition of PI3K. Crystal structures of BKM120 and derivatives in complex with tubulin and PI3K provide insights into the selective mode of action of this class of drugs. Our results raise concerns over BKM120's generally accepted mode of action, and provide a unique mechanistic basis for next-generation PI3K inhibitors with improved safety profiles and flexibility for use in combination therapies.
- 33Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C.; Mestan, J.; Hebeisen, P.; Hillmann, P.; Giese, B.; Zvelebil, M.; Fabbro, D.; Williams, R. L.; Rageot, D.; Wymann, M. P. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan-class I PI3K/mTOR inhibitor as clinical candidate in oncology. J. Med. Chem. 2017, 60 (17), 7524– 7538, DOI: 10.1021/acs.jmedchem.7b00930Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKhurrP&md5=54d4db75f6a8bda3f12eaf1d196352fd5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a Potent, Brain-Penetrant, Orally Bioavailable, Pan-Class I PI3K/mTOR Inhibitor as Clinical Candidate in OncologyBeaufils, Florent; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Bohnacker, Thomas; Melone, Anna; Marone, Romina; Jackson, Eileen; Zhang, Xuxiao; Sele, Alexander; Borsari, Chiara; Mestan, Jurgen; Hebeisen, Paul; Hillmann, Petra; Giese, Bernd; Zvelebil, Marketa; Fabbro, Doriano; Williams, Roger L.; Rageot, Denise; Wymann, Matthias P.Journal of Medicinal Chemistry (2017), 60 (17), 7524-7538CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphoinositide 3-kinase (PI3K) is deregulated in a wide variety of human tumors and triggers activation of protein kinase B (PKB/Akt) and mammalian target of rapamycin (mTOR). Here we describe the preclin. characterization of compd. 1 (PQR309, bimiralisib), a potent 4,6-dimorpholino-1,3,5-triazine-based pan-class I PI3K inhibitor, which targets mTOR kinase in a balanced fashion at higher concns. No off-target interactions were detected for 1 in a wide panel of protein kinase, enzyme, and receptor ligand assays. Moreover, 1 did not bind tubulin, which was obsd. for the structurally related 4 (BKM120, buparlisib). Compd. 1 is orally available, crosses the blood-brain barrier, and displayed favorable pharmacokinetic parameters in mice, rats, and dogs. Compd. 1 demonstrated efficiency in inhibiting proliferation in tumor cell lines and a rat xenograft model. This, together with the compd.'s safety profile, identifies 1 as a clin. candidate with a broad application range in oncol., including treatment of brain tumors or CNS metastasis. Compd. 1 is currently in phase II clin. trials for advanced solid tumors and refractory lymphoma.
- 34Tarantelli, C.; Gaudio, E.; Arribas, A. J.; Kwee, I.; Hillmann, P.; Rinaldi, A.; Cascione, L.; Spriano, F.; Bernasconi, E.; Guidetti, F.; Carrassa, L.; Pittau, R. B.; Beaufils, F.; Ritschard, R.; Rageot, D.; Sele, A.; Dossena, B.; Rossi, F. M.; Zucchetto, A.; Taborelli, M.; Gattei, V.; Rossi, D.; Stathis, A.; Stussi, G.; Broggini, M.; Wymann, M. P.; Wicki, A.; Zucca, E.; Cmiljanovic, V.; Fabbro, D.; Bertoni, F. PQR309 is a novel dual PI3K/mTOR inhibitor with preclinical antitumor activity in lymphomas as a single agent and in combination therapy. Clin. Cancer Res. 2018, 24 (1), 120– 129, DOI: 10.1158/1078-0432.CCR-17-1041Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVWktw%253D%253D&md5=730305c33b7f669b0eedd0614edd886cPQR309 Is a Novel Dual PI3K/mTOR Inhibitor with Preclinical Antitumor Activity in Lymphomas as a Single Agent and in Combination TherapyTarantelli, Chiara; Gaudio, Eugenio; Arribas, Alberto J.; Kwee, Ivo; Hillmann, Petra; Rinaldi, Andrea; Cascione, Luciano; Spriano, Filippo; Bernasconi, Elena; Guidetti, Francesca; Carrassa, Laura; Pittau, Roberta Bordone; Beaufils, Florent; Ritschard, Reto; Rageot, Denise; Sele, Alexander; Dossena, Barbara; Rossi, Francesca Maria; Zucchetto, Antonella; Taborelli, Monica; Gattei, Valter; Rossi, Davide; Stathis, Anastasios; Stussi, Georg; Broggini, Massimo; Wymann, Matthias P.; Wicki, Andreas; Zucca, Emanuele; Cmiljanovic, Vladimir; Fabbro, Doriano; Bertoni, FrancescoClinical Cancer Research (2018), 24 (1), 120-129CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Activation of the PI3K/mTOR signaling pathway is recurrent in different lymphoma types, and pharmacol. inhibition of the PI3K/mTOR pathway has shown activity in lymphoma patients. Here, we extensively characterized the in vitro and in vivo activity and the mechanism of action of PQR309 (bimiralisib), a novel oral selective dual PI3K/mTOR inhibitor under clin. evaluation, in preclin. lymphoma models. Exptl. Design: This study included preclin. in vitro activity screening on a large panel of cell lines, both as single agent and in combination, validation expts. on in vivo models and primary cells, proteomics and gene-expression profiling, and comparison with other signaling inhibitors. Results: PQR309 had in vitro antilymphoma activity as single agent and in combination with venetoclax, panobinostat, ibrutinib, lenalidomide, ARV-825, marizomib, and rituximab. Sensitivity to PQR309 was assocd. with specific baseline gene-expression features, such as high expression of transcripts coding for the BCR pathway. Combining proteomics and RNA profiling, we identified the different contribution of PQR309-induced protein phosphorylation and gene expression changes to the drug mechanism of action. Gene-expression signatures induced by PQR309 and by other signaling inhibitors largely overlapped. PQR309 showed activity in cells with primary or secondary resistance to idelalisib. Conclusions: On the basis of these results, PQR309 appeared as a novel and promising compd. that is worth developing in the lymphoma setting. Clin Cancer Res; 24(1); 120-9. ©2017 AACR.
- 35Wicki, A.; Brown, N.; Xyrafas, A.; Bize, V.; Hawle, H.; Berardi, S.; Cmiljanovic, N.; Cmiljanovic, V.; Stumm, M.; Dimitrijevic, S.; Herrmann, R.; Pretre, V.; Ritschard, R.; Tzankov, A.; Hess, V.; Childs, A.; Hierro, C.; Rodon, J.; Hess, D.; Joerger, M.; von Moos, R.; Sessa, C.; Kristeleit, R. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13). Eur. J. Cancer 2018, 96, 6– 16, DOI: 10.1016/j.ejca.2018.03.012Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Snu70%253D&md5=acbdfdb59498f5543ba7f02ed9709d0eFirst-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13)Wicki, Andreas; Brown, Nicholas; Xyrafas, Alexandros; Bize, Vincent; Hawle, Hanne; Berardi, Simona; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Stumm, Michael; Dimitrijevic, Sasa; Herrmann, Richard; Pretre, Vincent; Ritschard, Reto; Tzankov, Alexandar; Hess, Viviane; Childs, Alexa; Hierro, Cinta; Rodon, Jordi; Hess, Dagmar; Joerger, Markus; von Moos, Roger; Sessa, Cristiana; Kristeleit, RebeccaEuropean Journal of Cancer (2018), 96 (), 6-16CODEN: EJCAEL; ISSN:0959-8049. (Elsevier Ltd.)PQR309 is an orally bioavailable, balanced pan-phosphatidylinositol-3-kinase (PI3K), mammalian target of rapamycin (mTOR) C1 and mTORC2 inhibitor. This is an accelerated titrn., 3 D 3 dose-escalation, open-label phase Itrial of continuous once-daily (OD) PQR309 administration to evaluate the safety, pharmacokinetics (PK) and pharmacodynamics in patients with advanced solid tumors. Primary objectives were to det. the max. tolerated dose (MTD) and recommended phase 2 dose (RP2D).Twenty-eight patients were included in six dosing cohorts and treated at a daily PQR309 dose ranging from 10 to 150 mg. Common adverse events (AEs; ≥30% patients) included fatigue, hyperglycemia, nausea, diarrhea, constipation, rash, anorexia and vomiting. Grade (G) 3 or 4 drug-related AEs were seen in 13 (46%) and three (11%) patients, resp. Dose-limiting toxicity (DLT) was obsd. in two patients at 100 mg OD (>14-d interruption in PQR309 due to G3 rash, G2 hyperbilirubinemia, G4 suicide attempt; dose redn. due to G3 fatigue, G2 diarrhoea, G4 transaminitis) and one patient at 80 mg (G3 hyperglycemia >7 d). PK shows fast absorption (Tmax 1-2 h) and dose proportionality for Cmax and area under the curve. A partial response in a patient with metastatic thymus cancer, 24% disease vol. redn. in a patient with sinonasal cancer and stable disease for more than 16 wk in a patient with clear cell Bartholin's gland cancer were obsd.The MTD and RP2D of PQR309 is 80 mg of orally OD. PK is dose-proportional. PD shows PI3K pathway phosphoprotein downregulation in paired tumor biopsies. Clin. activity was obsd. in patients with and without PI3K pathway dysregulation.
- 36Fang, Z.; Song, Y.; Zhan, P.; Zhang, Q.; Liu, X. Conformational restriction: an effective tactic in “follow-on”-based drug discovery. Future Med. Chem. 2014, 6 (8), 885– 901, DOI: 10.4155/fmc.14.50Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVemsrzE&md5=dcf5fa6cead84c77b3603c7e0800f49bConformational restriction: an effective tactic in 'follow-on'-based drug discoveryFang, Zengjun; Song, Yu'ning; Zhan, Peng; Zhang, Qingzhu; Liu, XinyongFuture Medicinal Chemistry (2014), 6 (8), 885-901CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. The conformational restriction (rigidification) of a flexible ligand has often been a commonly used strategy in drug design, as it can minimize the entropic loss assocd. with the ligand adopting a preferred conformation for binding, which leads to enhanced potency for a given physiol. target, improved selectivity for isoforms and reduced the possibility of drug metab. Therefore, the application of conformational restriction strategy is a core aspect of drug discovery and development that is widely practiced by medicinal chemists either deliberately or subliminally. The present review will highlight current representative examples and a brief overview on the rational design of conformationally restricted agents as well as discuss its advantages over the flexible counterparts.
- 37Cmiljanovic, V.; Hebeisen, P.; Jackson, E.; Beaufils, F.; Bohnacker, T.; Wymann, M. P. Conformationally Restricted PI3K and mTOR Inhibitors. Patent WO2015049369, 2015.Google ScholarThere is no corresponding record for this reference.
- 38Leroux, F. R.; Manteau, B.; Vors, J. P.; Pazenok, S. Trifluoromethyl ethers--synthesis and properties of an unusual substituent. Beilstein J. Org. Chem. 2008, 4 (13), DOI: 10.3762/bjoc.4.13 .Google ScholarThere is no corresponding record for this reference.
- 39Burger, M. T.; Pecchi, S.; Wagman, A.; Ni, Z. J.; Knapp, M.; Hendrickson, T.; Atallah, G.; Pfister, K.; Zhang, Y.; Bartulis, S.; Frazier, K.; Ng, S.; Smith, A.; Verhagen, J.; Haznedar, J.; Huh, K.; Iwanowicz, E.; Xin, X.; Menezes, D.; Merritt, H.; Lee, I.; Wiesmann, M.; Kaufman, S.; Crawford, K.; Chin, M.; Bussiere, D.; Shoemaker, K.; Zaror, I.; Maira, S. M.; Voliva, C. F. Identification of NVP-BKM120 as a potent, selective, orally bioavailable class I PI3 Kinase inhibitor for treating cancer. ACS Med. Chem. Lett. 2011, 2 (10), 774– 779, DOI: 10.1021/ml200156tGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVOktrrI&md5=5d00d73dda1fc0d8f0f07d12900282edIdentification of NVP-BKM120 as a Potent, Selective, Orally Bioavailable Class I PI3 Kinase Inhibitor for Treating CancerBurger, Matthew T.; Pecchi, Sabina; Wagman, Allan; Ni, Zhi-Jie; Knapp, Mark; Hendrickson, Thomas; Atallah, Gordana; Pfister, Keith; Zhang, Yanchen; Bartulis, Sarah; Frazier, Kelly; Ng, Simon; Smith, Aaron; Verhagen, Joelle; Haznedar, Joshua; Huh, Kay; Iwanowicz, Ed; Xin, Xiaohua; Menezes, Daniel; Merritt, Hanne; Lee, Isabelle; Wiesmann, Marion; Kaufman, Susan; Crawford, Kenneth; Chin, Michael; Bussiere, Dirksen; Shoemaker, Kevin; Zaror, Isabel; Maira, Sauveur-Michel; Voliva, Charles F.ACS Medicinal Chemistry Letters (2011), 2 (10), 774-779CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Phosphoinositide-3-kinases (PI3Ks) are important oncol. targets due to the deregulation of this signaling pathway in a wide variety of human cancers. Herein we describe the structure guided optimization of a series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines where the pharmacokinetic properties were improved by modulating the electronics of the 6-position heterocycle, and the overall druglike properties were fine-tuned further by modification of the 4-position substituent. The resulting 2,4-bismorpholino 6-heterocyclic pyrimidines are potent class I PI3K inhibitors showing mechanism modulation in PI3K dependent cell lines and in vivo efficacy in tumor xenograft models with PI3K pathway deregulation (A2780 ovarian and U87MG glioma). These efforts culminated in the discovery of 15 (NVP-BKM120), currently in Phase II clin. trials for the treatment of cancer.
- 40Rousseau, J. F.; Chekroun, I.; Ferey, V.; Labrosse, J. R. Concise preparation of a stable cyclic sulfamidate intermediate in the synthesis of a enantiopure chiral active diamine derivative. Org. Process Res. Dev. 2015, 19 (4), 506– 513, DOI: 10.1021/op500264vGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXksVOrurk%253D&md5=72c5ea832b893e2bccf01a80c9ea1734Concise Preparation of a Stable Cyclic Sulfamidate Intermediate in the Synthesis of a Enantiopure Chiral Active Diamine DerivativeRousseau, Jean-Francois; Chekroun, Isaac; Ferey, Vincent; Labrosse, Jean RobertOrganic Process Research & Development (2015), 19 (4), 506-513CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A potentially scalable route to the nonracemic antipsychotic candidate SSR 504374 I was developed using the stereoselective substitution reaction of nonracemic sulfamidate II with 2-chloro-3-trifluoromethylbenzamide as the key step. II was prepd. in seven steps from 2-benzoylpyridine by chemo- and diastereoselective hydrogenation, sepn. of the desired racemic erythro diastereomer, resoln. with di-p-toluoyl-(+)-tartaric acid, formation of a sulfamidite, and oxidn. at sulfur with RuCl3 and sodium hypochlorite; a workup for the sulfamidite oxidn. using isopropanol was developed to avoid darkening of the intermediate due to oxidn. by RuO2 and RuO4 left in the sulfamidate product.
- 41Brown, G. R.; Foubister, A. J.; Wright, B. Chiral synthesis of 3-substituted morpholines via serine enantiomers and reductions of 5-oxomorpholine-3-carboxylates. J. Chem. Soc., Perkin Trans. 1 1985, 1, 2577– 2580, DOI: 10.1039/p19850002577Google ScholarThere is no corresponding record for this reference.
- 42Hebeisen, P.; Alker, A.; Buerkler, M. Iterative one pot reactions of a chiral sulfamidate with 2,4,6-trichloropyridine: regiocontrolled synthesis of linear and angular chiral dipyrrolidino pyridines. Heterocycles 2012, 85 (1), 65– 72, DOI: 10.3987/COM-11-12360Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpslWnug%253D%253D&md5=07450591f843628878b047a25205e5bfIterative one pot reactions of a chiral sulfamidate with 2,4,6-trichloropyridine: Regiocontrolled synthesis of linear and angular chiral dipyrrolidino pyridinesHebeisen, Paul; Alker, Andre; Buerkler, MarkusHeterocycles (2012), 85 (1), 65-72CODEN: HTCYAM; ISSN:0385-5414. (Japan Institute of Heterocyclic Chemistry)The product of the ring opening of a chiral sulfamidate with the 3-lithiopyridine species obtained by deprotonation of 2,4,6-trichloropyridine with BuLi was deprotonated again in situ with BuLi and reacted with a 2nd equiv. of the sulfamidate furnishing a bis(β-aminoethyl)pyridine deriv., which could be cyclized regioselectively to linear or angular chiral dipyrrolidinopyridines.
- 43Zask, A.; Kaplan, J.; Verheijen, J. C.; Richard, D. J.; Curran, K.; Brooijmans, N.; Bennett, E. M.; Toral-Barza, L.; Hollander, I.; Ayral-Kaloustian, S.; Yu, K. Morpholine derivatives greatly enhance the selectivity of mammalian target of rapamycin (mTOR) inhibitors. J. Med. Chem. 2009, 52 (24), 7942– 7945, DOI: 10.1021/jm901415xGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVWrtb3J&md5=5ef1eee5aa79531970049d3d26b46eb2Morpholine Derivatives Greatly Enhance the Selectivity of Mammalian Target of Rapamycin (mTOR) InhibitorsZask, Arie; Kaplan, Joshua; Verheijen, Jeroen C.; Richard, David J.; Curran, Kevin; Brooijmans, Natasja; Bennett, Eric M.; Toral-Barza, Lourdes; Hollander, Irwin; Ayral-Kaloustian, Semiramis; Yu, KerJournal of Medicinal Chemistry (2009), 52 (24), 7942-7945CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Dramatic improvements in mTOR-targeting selectivity were achieved by replacing morpholine in pyrazolopyrimidine inhibitors with bridged morpholines. Analogs I [R1 = (R)- or (S)-3-methyl-4-morpholinyl, 2-methyl-4-morpholinyl, 8-oxa-3-azabicyclo[3.2.1]octan-3-yl, etc.; R2 = Me, Et, cyclopropyl, FCH2CH2, 3-pyridyl, 4-pyridyl; R3 = F3CCH2, 1-methoxycarbonyl-4-piperidinyl, 1-ethoxycarbonyl-4-piperidinyl, 3-pyridylmethyl] with subnanomolar mTOR IC50 values and up to 26000-fold selectivity vs. PI3Kα were prepd. Chiral morpholines gave inhibitors whose enantiomers had different selectivity and potency profiles. Mol. modeling suggests that a single amino acid difference between PI3K and mTOR (Phe961Leu) accounts for the profound selectivity seen by creating a deeper pocket in mTOR that can accommodate bridged morpholines.
- 44Thomas, V. H.; Bhattachar, S.; Hitchingham, L.; Zocharski, P.; Naath, M.; Surendran, N.; Stoner, C. L.; El-Kattan, A. The road map to oral bioavailability: an industrial perspective. Expert Opin. Drug Metab. Toxicol. 2006, 2 (4), 591– 608, DOI: 10.1517/17425255.2.4.591Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnt1ahsLs%253D&md5=79cf4cea1b89914bd3a8826b5d7360f9The road map to oral bioavailability: an industrial perspectiveThomas, V. Hayden; Bhattachar, Shobha; Hitchingham, Linda; Zocharski, Philip; Naath, Maryanne; Surendran, Narayanan; Stoner, Chad L.; El-Kattan, AymanExpert Opinion on Drug Metabolism & Toxicology (2006), 2 (4), 591-608CODEN: EODMAP; ISSN:1742-5255. (Informa Healthcare)A review. Optimization of oral bioavailability is a continuing challenge for the pharmaceutical and biotechnol. industries. The no. of potential drug candidates requiring in vivo evaluation has significantly increased with the advent of combinatorial chem. In addn., drug discovery programs are increasingly forced into more lipophilic and lower soly. chem. space. To aid in the use of in vitro and in silico tools as well as reduce the no. of in vivo studies required, a team-based discussion tool is proposed that provides a road map' to guide the selection of profiling assays that should be considered when optimizing oral bioavailability. This road map divides the factors that contribute to poor oral bioavailability into two interrelated categories: absorption and metab. This road map provides an interface for cross discipline discussions and a systematic approach to the experimentation that drives the drug discovery process towards a common goal - acceptable oral bioavailability using minimal resources in an acceptable time frame.
- 45Mortensen, D. S.; Fultz, K. E.; Xu, S.; Xu, W.; Packard, G.; Khambatta, G.; Gamez, J. C.; Leisten, J.; Zhao, J.; Apuy, J.; Ghoreishi, K.; Hickman, M.; Narla, R. K.; Bissonette, R.; Richardson, S.; Peng, S. X.; Perrin-Ninkovic, S.; Tran, T.; Shi, T.; Yang, W. Q.; Tong, Z.; Cathers, B. E.; Moghaddam, M. F.; Canan, S. S.; Worland, P.; Sankar, S.; Raymon, H. K. CC-223, a potent and selective inhibitor of mTOR kinase: in vitro and in vivo characterization. Mol. Cancer Ther. 2015, 14 (6), 1295– 1305, DOI: 10.1158/1535-7163.MCT-14-1052Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpsFOgurk%253D&md5=f33b21dc89dedaf14fab54a819afb917CC-223, a Potent and Selective Inhibitor of mTOR Kinase: In Vitro and In Vivo CharacterizationMortensen, Deborah S.; Fultz, Kimberly E.; Xu, Shuichan; Xu, Weiming; Packard, Garrick; Khambatta, Godrej; Gamez, James C.; Leisten, Jim; Zhao, Jingjing; Apuy, Julius; Ghoreishi, Kamran; Hickman, Matt; Narla, Rama Krishna; Bissonette, Rene; Richardson, Samantha; Peng, Sophie X.; Perrin-Ninkovic, Sophie; Tran, Tam; Shi, Tao; Yang, Wen Qing; Tong, Zeen; Cathers, Brian E.; Moghaddam, Mehran F.; Canan, Stacie S.; Worland, Peter; Sankar, Sabita; Raymon, Heather K.Molecular Cancer Therapeutics (2015), 14 (6), 1295-1305CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)MTOR is a serine/threonine kinase that regulates cell growth, metab., proliferation, and survival. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are crit. mediators of the PI3K-AKT pathway, which is frequently mutated in many cancers, leading to hyperactivation of mTOR signaling. Although rapamycin analogs, allosteric inhibitors that target only the mTORC1 complex, have shown some clin. activity, it is hypothesized that mTOR kinase inhibitors, blocking both mTORC1 and mTORC2 signaling, will have expanded therapeutic potential. Here, we describe the preclin. characterization of CC-223. CC-223 is a potent, selective, and orally bioavailable inhibitor of mTOR kinase, demonstrating inhibition of mTORC1 (pS6RP and p4EBP1) and mTORC2 [pAKT(S473)] in cellular systems. Growth inhibitory activity was demonstrated in hematol. and solid tumor cell lines. mTOR kinase inhibition in cells, by CC-223, resulted in more complete inhibition of the mTOR pathway biomarkers and improved antiproliferative activity as compared with rapamycin. Growth inhibitory activity and apoptosis was demonstrated in a panel of hematol. cancer cell lines. Correlative anal. revealed that IRF4 expression level assocs. with resistance, whereas mTOR pathway activation seems to assoc. with sensitivity. Treatment with CC-223 afforded in vivo tumor biomarker inhibition in tumor-bearing mice, after a single oral dose. CC-223 exhibited dose-dependent tumor growth inhibition in multiple solid tumor xenografts. Significant inhibition of mTOR pathway markers pS6RP and pAKT in CC-223-treated tumors suggests that the obsd. antitumor activity of CC-223 was mediated through inhibition of both mTORC1 and mTORC2. CC-223 is currently in phase I clin. trials. Mol Cancer Ther; 14(6); 1295-305. ©2015 AACR.
- 46Nosik, P. S.; Ryabukhin, S. V.; Artamonov, O. S.; Grygorenko, O. O. Synthesis of trans-disubstituted pyrazolylcyclopropane building blocks. Monatsh. Chem. 2016, 147 (9), 1629– 1636, DOI: 10.1007/s00706-016-1726-6Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmsFyksbg%253D&md5=1b4cd76fed0aa27fc3134d8bbe4a301fSynthesis of trans-disubstituted pyrazolylcyclopropane building blocksNosik, Pavel S.; Ryabukhin, Sergey V.; Artamonov, Oleksiy S.; Grygorenko, Oleksandr O.Monatshefte fuer Chemie (2016), 147 (9), 1629-1636CODEN: MOCMB7; ISSN:0026-9247. (Springer-Verlag GmbH)Diastereoselective synthesis of trans-disubstituted pyrazolylcyclopropane building blocks (i.e. carboxylic acids and amines) is described starting from easily available pyrazolecarbaldehydes. The key step of the synthesis was Corey-Chaikowsky cyclopropanation of the corresponding α,β-unsatd. Weinreb amides. The title compds. were prepd. in four or six steps and 32-60 and 17-40 % overall yields, resp., on up to 50 g scale. The building blocks obtained are good starting points for the design of lead-like libraries of peptidomimetic drugs.
- 47Zhang, L.; Luo, S.; Mi, X.; Liu, S.; Qiao, Y.; Xu, H.; Cheng, J. P. Combinatorial synthesis of functionalized chiral and doubly chiral ionic liquids and their applications as asymmetric covalent/non-covalent bifunctional organocatalysts. Org. Biomol. Chem. 2008, 6 (3), 567– 576, DOI: 10.1039/B713843AGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtVWhs7o%253D&md5=4cec4a7bff2e3b65c7aac57156230d27Combinatorial synthesis of functionalized chiral and doubly chiral ionic liquids and their applications as asymmetric covalent/non-covalent bifunctional organocatalystsZhang, Long; Luo, Sanzhong; Mi, Xueling; Liu, Song; Qiao, Yupu; Xu, Hui; Cheng, Jin-PeiOrganic & Biomolecular Chemistry (2008), 6 (3), 567-576CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A facile combinatorial strategy was developed for the construction of libraries of functionalized chiral ionic liqs. (FCILs) including doubly chiral ionic liqs. and bis-functional chiral ionic liqs. These FCIL libraries have the potential to be used as asym. catalysts or chiral ligands. As an example, novel asym. bifunctional catalysts were developed by simultaneously incorporating functional groups onto the cation and anion. The resultant bis-functionalized CILs showed significantly improved stereoselectivity over the mono-functionalized parent CILs.
- 48Fabian, M. A.; Biggs, W. H.; Treiber, D. K.; Atteridge, C. E.; Azimioara, M. D.; Benedetti, M. G.; Carter, T. A.; Ciceri, P.; Edeen, P. T.; Floyd, M.; Ford, J. M.; Galvin, M.; Gerlach, J. L.; Grotzfeld, R. M.; Herrgard, S.; Insko, D. E.; Insko, M. A.; Lai, A. G.; Lélias, J. M.; Mehta, S. A.; Milanov, Z. V.; Velasco, A. M.; Wodicka, L. M.; Patel, H. K.; Zarrinkar, P. P.; Lockhart, D. J. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat. Biotechnol. 2005, 23, 329– 336, DOI: 10.1038/nbt1068Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXitF2nt7w%253D&md5=6aedd5ceb8f77cd26ee50425dcec7bdfA small molecule-kinase interaction map for clinical kinase inhibitorsFabian, Miles A.; Biggs, William H.; Treiber, Daniel K.; Atteridge, Corey E.; Azimioara, Mihai D.; Benedetti, Michael G.; Carter, Todd A.; Ciceri, Pietro; Edeen, Philip T.; Floyd, Mark; Ford, Julia M.; Galvin, Margaret; Gerlach, Jay L.; Grotzfeld, Robert M.; Herrgard, Sanna; Insko, Darren E.; Insko, Michael A.; Lai, Andiliy G.; Lelias, Jean-Michel; Mehta, Shamal A.; Milanov, Zdravko V.; Velasco, Anne Marie; Wodicka, Lisa M.; Patel, Hitesh K.; Zarrinkar, Patrick P.; Lockhart, David J.Nature Biotechnology (2005), 23 (3), 329-336CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Kinase inhibitors show great promise as a new class of therapeutics. Here the authors describe an efficient way to det. kinase inhibitor specificity by measuring binding of small mols. to the ATP site of kinases. The authors have profiled 20 kinase inhibitors, including 16 that are approved drugs or in clin. development, against a panel of 119 protein kinases. The authors find that specificity varies widely and is not strongly correlated with chem. structure or the identity of the intended target. Many novel interactions were identified, including tight binding of the p38 inhibitor BIRB-796 to an imatinib-resistant variant of the ABL kinase, and binding of imatinib to the SRC-family kinase LCK. The authors also show that mutations in the epidermal growth factor receptor (EGFR) found in gefitinib-responsive patients do not affect the binding affinity of gefitinib or erlotinib. Our results represent a systematic small mol.-protein interaction map for clin. compds. across a large no. of related proteins.
- 49Karaman, M. W.; Herrgard, S.; Treiber, D. K.; Gallant, P.; Atteridge, C. E.; Campbell, B. T.; Chan, K. W.; Ciceri, P.; Davis, M. I.; Edeen, P. T.; Faraoni, R.; Floyd, M.; Hunt, J. P.; Lockhart, D. J.; Milanov, Z. V.; Morrison, M. J.; Pallares, G.; Patel, H. K.; Pritchard, S.; Wodicka, L. M.; Zarrinkar, P. P. A quantitative analysis of kinase inhibitor selectivity. Nat. Biotechnol. 2008, 26, 127– 132, DOI: 10.1038/nbt1358Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXisFGlsQ%253D%253D&md5=346265d412853ced636ad4128ed8a76fA quantitative analysis of kinase inhibitor selectivityKaraman, Mazen W.; Herrgard, Sanna; Treiber, Daniel K.; Gallant, Paul; Atteridge, Corey E.; Campbell, Brian T.; Chan, Katrina W.; Ciceri, Pietro; Davis, Mindy I.; Edeen, Philip T.; Faraoni, Raffaella; Floyd, Mark; Hunt, Jeremy P.; Lockhart, Daniel J.; Milanov, Zdravko V.; Morrison, Michael J.; Pallares, Gabriel; Patel, Hitesh K.; Pritchard, Stephanie; Wodicka, Lisa M.; Zarrinkar, Patrick P.Nature Biotechnology (2008), 26 (1), 127-132CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Kinase inhibitors are a new class of therapeutics with a propensity to inhibit multiple targets. The biol. consequences of multikinase activity are poorly defined, and an important step toward understanding the relationship between selectivity, efficacy and safety is the exploration of how inhibitors interact with the human kinome. The authors present interaction maps for 38 kinase inhibitors across a panel of 317 kinases representing >50% of the predicted human protein kinome. The data constitute the most comprehensive study of kinase inhibitor selectivity to date and reveal a wide diversity of interaction patterns. To enable a global anal. of the results, the authors introduce the concept of a selectivity score as a general tool to quantify and differentiate the obsd. interaction patterns. The authors further investigate the impact of panel size and find that small assay panels do not provide a robust measure of selectivity.
Cited By
This article is cited by 22 publications.
- Jindi Huang, Liye Chen, Jiangxia Wu, Daiqiao Ai, Ji-Quan Zhang, Tie-Gen Chen, Ling Wang. Targeting the PI3K/AKT/mTOR Signaling Pathway in the Treatment of Human Diseases: Current Status, Trends, and Solutions. Journal of Medicinal Chemistry 2022, 65
(24)
, 16033-16061. https://doi.org/10.1021/acs.jmedchem.2c01070
- Chiara Borsari, Erhan Keles, Denise Rageot, Andrea Treyer, Thomas Bohnacker, Lukas Bissegger, Martina De Pascale, Anna Melone, Rohitha Sriramaratnam, Florent Beaufils, Matthias Hamburger, Paul Hebeisen, Wolfgang Löscher, Doriano Fabbro, Petra Hillmann, Matthias P. Wymann. 4-(Difluoromethyl)-5-(4-((3R,5S)-3,5-dimethylmorpholino)-6-((R)-3-methylmorpholino)-1,3,5-triazin-2-yl)pyridin-2-amine (PQR626), a Potent, Orally Available, and Brain-Penetrant mTOR Inhibitor for the Treatment of Neurological Disorders. Journal of Medicinal Chemistry 2020, 63
(22)
, 13595-13617. https://doi.org/10.1021/acs.jmedchem.0c00620
- Simone Bonazzi, Carleton P. Goold, Audrey Gray, Noel M. Thomsen, Jill Nunez, Rajeshri G. Karki, Aakruti Gorde, Jonathan D. Biag, Hasnain A. Malik, Yingchuan Sun, Guiqing Liang, Danuta Lubicka, Sarah Salas, Nancy Labbe-Giguere, Erin P. Keaney, Stephanie McTighe, Shanming Liu, Lin Deng, Grazia Piizzi, Franco Lombardo, Doug Burdette, Jean-Cosme Dodart, Christopher J. Wilson, Stefan Peukert, Daniel Curtis, Lawrence G. Hamann, Leon O. Murphy. Discovery of a Brain-Penetrant ATP-Competitive Inhibitor of the Mechanistic Target of Rapamycin (mTOR) for CNS Disorders. Journal of Medicinal Chemistry 2020, 63
(3)
, 1068-1083. https://doi.org/10.1021/acs.jmedchem.9b01398
- Denise Rageot, Florent Beaufils, Chiara Borsari, Alix Dall’Asen, Markus Neuburger, Paul Hebeisen, Matthias P. Wymann. Scalable, Economical, and Practical Synthesis of 4-(Difluoromethyl)pyridin-2-amine, a Key Intermediate for Lipid Kinase Inhibitors. Organic Process Research & Development 2019, 23
(11)
, 2416-2424. https://doi.org/10.1021/acs.oprd.9b00312
- Chiara Borsari, Denise Rageot, Florent Beaufils, Thomas Bohnacker, Erhan Keles, Ivan Buslov, Anna Melone, Alexander M. Sele, Paul Hebeisen, Doriano Fabbro, Petra Hillmann, Matthias P. Wymann. Preclinical Development of PQR514, a Highly Potent PI3K Inhibitor Bearing a Difluoromethyl–Pyrimidine Moiety. ACS Medicinal Chemistry Letters 2019, 10
(10)
, 1473-1479. https://doi.org/10.1021/acsmedchemlett.9b00333
- Ahmed H.E. Hassan, Mohammad Maqusood Alam, Trong-Nhat Phan, Kyung-Hwa Baek, Hyeryon Lee, Soo Bin Cho, Chae Hyeon Lee, Yeon Ju Kim, Joo Hwan No, Yong Sup Lee. Repurposing of conformationally-restricted cyclopentane-based AKT–inhibitors leads to discovery of potential and more selective antileishmanial agents than miltefosine. Bioorganic Chemistry 2023, 141 , 106890. https://doi.org/10.1016/j.bioorg.2023.106890
- Shashwati Paul, Daniel Adelfinsky, Christophe Salome, Thomas Fessard, M. Kevin Brown. 2,5-disubstituted bicyclo[2.1.1]hexanes as rigidified cyclopentane variants. Chemical Science 2023, 14
(30)
, 8070-8075. https://doi.org/10.1039/D3SC02695G
- Sara Asadi, Hossein Mehrabi. Efficient synthesis of some novel pyrimido[4,5‐
e
][1,3]oxazine dione derivatives via one‐pot three‐component reaction under thermal and thermal microwave‐assisted conditions. Journal of Heterocyclic Chemistry 2023, 60
(3)
, 497-503. https://doi.org/10.1002/jhet.4605
- Martina De Pascale, Lukas Bissegger, Chiara Tarantelli, Florent Beaufils, Alessandro Prescimone, Hayget Mohamed Seid Hedad, Omar Kayali, Clara Orbegozo, Luka Raguž, Thorsten Schaefer, Paul Hebeisen, Francesco Bertoni, Matthias P. Wymann, Chiara Borsari. Investigation of morpholine isosters for the development of a potent, selective and metabolically stable mTOR kinase inhibitor. European Journal of Medicinal Chemistry 2023, 248 , 115038. https://doi.org/10.1016/j.ejmech.2022.115038
- Uros Stojiljkovic, Claudio Meyer, Pierre Boulay, Paul Hebeisen, Denise Rageot, Matthias P. Wymann, Chiara Borsari. Stereospecific Synthesis of Substituted Sulfamidates as Privileged Morpholine Building Blocks. Synthesis 2023, 55
(03)
, 499-509. https://doi.org/10.1055/a-1915-7794
- Patrik Oleksak, Eugenie Nepovimova, Zofia Chrienova, Kamil Musilek, Jiri Patocka, Kamil Kuca. Contemporary mTOR inhibitor scaffolds to diseases breakdown: A patent review (2015–2021). European Journal of Medicinal Chemistry 2022, 238 , 114498. https://doi.org/10.1016/j.ejmech.2022.114498
- Beibei Mao, Qi Zhang, Li Ma, Dong-Sheng Zhao, Pan Zhao, Peizheng Yan. Overview of Research into mTOR Inhibitors. Molecules 2022, 27
(16)
, 5295. https://doi.org/10.3390/molecules27165295
- Bohdan Vashchenko, Oleksandr Grygorenko, Oleksandr Stepaniuk. Heterocyclizations of β-alkoxy, β-diaminoalkyl, and related
β-functionalized enones (enals) with NCN-binucleophiles. Ukrainica Bioorganica Acta 2022, 17
(1)
, 56-71. https://doi.org/10.15407/bioorganica2022.01.056
- Chiara Borsari, Matthias P. Wymann. Targeting Phosphoinositide 3-Kinase – Five Decades of Chemical Space Exploration. CHIMIA 2021, 75
(12)
, 1037. https://doi.org/10.2533/chimia.2021.1037
- Chiara Borsari, Martina De Pascale, Matthias P. Wymann. Chemical and Structural Strategies to Selectively Target mTOR Kinase. ChemMedChem 2021, 16
(18)
, 2744-2759. https://doi.org/10.1002/cmdc.202100332
- Li Chen, Rong Huang, Xing-Han Yun, Tian-Hui Hao, Sheng-Jiao Yan. Multi-component cascade reaction of 3-formylchromones: highly selective synthesis of 4,5-dihydro-[4,5′-bipyrimidin]-6(1
H
)-one derivatives. Chemical Communications 2021, 57
(62)
, 7657-7660. https://doi.org/10.1039/D1CC02437J
- Chiara Borsari, Erhan Keles, Andrea Treyer, Martina De Pascale, Paul Hebeisen, Matthias Hamburger, Matthias P. Wymann. Second-generation tricyclic pyrimido-pyrrolo-oxazine mTOR inhibitor with predicted blood–brain barrier permeability. RSC Medicinal Chemistry 2021, 12
(4)
, 579-583. https://doi.org/10.1039/D0MD00408A
- Ruoyu He, Bingyong Xu, Li Ping, Xiaoqing Lv. Structural optimization towards promising β-methyl-4-acrylamido quinoline derivatives as PI3K/mTOR dual inhibitors for anti-cancer therapy: The in vitro and in vivo biological evaluation. European Journal of Medicinal Chemistry 2021, 214 , 113249. https://doi.org/10.1016/j.ejmech.2021.113249
- S. V. Fedoseev, O. E. Ershov. Reaction of 4-Halo-3-hydroxyfuro[3,4-c]pyridin-1(3H)-ones with Morpholine and Thiomorpholine. Russian Journal of Organic Chemistry 2021, 57
(3)
, 483-485. https://doi.org/10.1134/S1070428021030234
- Rajkumar Lalji Sahani, Raquel Diana-Rivero, Sanjeev Kumar V. Vernekar, Lei Wang, Haijuan Du, Huanchun Zhang, Andres Emanuelli Castaner, Mary C. Casey, Karen A. Kirby, Philip R. Tedbury, Jiashu Xie, Stefan G. Sarafianos, Zhengqiang Wang. Design, Synthesis and Characterization of HIV-1 CA-Targeting Small Molecules: Conformational Restriction of PF74. Viruses 2021, 13
(3)
, 479. https://doi.org/10.3390/v13030479
- Ting-Ting Wu, Qing-Qing Guo, Zi-Li Chen, Li-Li Wang, Yao Du, Rui Chen, Yuan-Hu Mao, Sheng-Gang Yang, Jing Huang, Jian-Ta Wang, Ling Wang, Lei Tang, Ji-Quan Zhang. Design, synthesis and bioevaluation of novel substituted triazines as potential dual PI3K/mTOR inhibitors. European Journal of Medicinal Chemistry 2020, 204 , 112637. https://doi.org/10.1016/j.ejmech.2020.112637
- Cedric Magaway, Eugene Kim, Estela Jacinto. Targeting mTOR and Metabolism in Cancer: Lessons and Innovations. Cells 2019, 8
(12)
, 1584. https://doi.org/10.3390/cells8121584
Abstract
Figure 1
Figure 1. Chemical structures of rapamycin and rapalogs and a selection of ATP-competitive mTOR kinase inhibitor (TORKi) compounds.
Figure 2
Figure 2. Strategy for the development of mTOR selective inhibitors starting from PQR309 (1): rigidification strategy (red dotted lines) and removal of trifluoromethyl group from the 2-aminopyridine moiety (blue).
Figure 3
Figure 3. (A) Docking of compound 3a (plum) into PI3Kγ (gray) starting from PDB code 5JHB (see ref (32)). Structural water molecules are shown in red, and water-mediated H-bonds are depicted as dashed black lines. (B) Docking of compound 2a (gold) and (C) compound 2b (green) into mTOR (turquoise) starting from PDB code 4JT6. The important features for mTOR selectivity are depicted in a ball and stick representation. (D) Docking of compound 2a (gold) and (E) compound 2b (green) into PI3Kα (gray) starting from PDB code 3ZIM. The exit vector from the restricted morpholine oxygen is shown as a black arrow.
Scheme 1
Scheme 1. c
aPrepared according to ref (32).
bPrepared according to procedure vii. After the reaction, the two regioisomers (26 and 32) were separated by column chromatography.
c(A) Reagents and conditions: (i) (1) benzaldehyde, 2 M NaOH, rt, 30 min; (2) NaBH4, 5 °C → rt, 1 h; (ii) (1) chloroacetyl chloride, K2CO3, THF/H2O, 0 °C, 1 h; (2) NaOH, 5 °C, 2 h; (iii) borane–dimethyl sulfide complex, Et3N, THF, 0 °C → 65 °C, 5 h; (iv) Pd/C, H2, 2.8 bar, 48 h; (v) thionyl chloride, imidazole, DCM, −5 °C → rt → 0 °C, 2 h; (vi) ruthenium(IV) oxide hydrate, NaIO4, rt, o/n. (B) Reagents and conditions: (vii) morpholine derivative (Mn–H), DIPEA, DCM, 0 °C → rt, o/n; (viii) (1) n-BuLi, CuI, −78 °C → rt, o/n; (2) HCl conc, MeOH, 45 °C, 4–6 h; (3) NaOH, H2O, rt, 1–16 h; (ix) (1) boronic ester 38 or 41, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 2–16 h; (2) HCl, dioxane/H2O, 60 °C, 3–16 h (for 2a, 2b, 2c, 2d, 6a, 6b, 7a, 7b, 8a, 8b, 9a, 9b, 10a, 10b, and 14b); (x) 2-aminopyridine-5-boronic acid pinacol ester, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C (for 11b), o/n; (xi) boronic ester 39, Pd(dppf)Cl2 (cat.), CsCO3, THF, Δ, o/n (for 12b); (xii) (1) boronic ester generated in situ, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 3–3.5 h; (2) HCl, 80 °C, o/n (for 13b and 15b); (xiii) (1) boronic ester generated in situ, XPhosPdG2 (cat.), K3PO4, dioxane/H2O, 95 °C, 2–16 h (isolated intermediates 18b and 19b); (2) 18b or 19b, TFA, DCM, 0 °C → rt, 1–3 h (for 16b and 17b).
Figure 4
Figure 4. Plasma and brain concentration of (A) compound 7b and (B) compound 12b after po dosing at 5 mg/kg in male Sprague Dawley rats. Stability of compound 11b (5 μM) with primary hepatocytes from (C) mice (green) and rats (turquoise) and (D) dogs (red) and humans (black) (n = 2). All values are the mean ± SEM. Error bars are not shown when smaller than the symbols.
Figure 5
Figure 5. Pie chart showing the percentage of compound 11b and its metabolites after 60 min of incubation with human recombinant (A) CYP1A1 and (B) CYP1A2. (C) Major metabolites observed upon incubation of 11b with CYP1A1.
References
ARTICLE SECTIONSThis article references 49 other publications.
- 1Wymann, M. P.; Schneiter, R. Lipid signalling in disease. Nat. Rev. Mol. Cell Biol. 2008, 9 (2), 162– 176, DOI: 10.1038/nrm2335Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXovFOgug%253D%253D&md5=c96aabfdfa8e93c9e98386f1a407de85Lipid signalling in diseaseWymann, Matthias P.; Schneiter, RogerNature Reviews Molecular Cell Biology (2008), 9 (2), 162-176CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Signaling lipids such as eicosanoids, phosphoinositides, sphingolipids and fatty acids control important cellular processes, including cell proliferation, apoptosis, metab. and migration. Extracellular signals from cytokines, growth factors and nutrients control the activity of a key set of lipid-modifying enzymes: phospholipases, prostaglandin synthase, 5-lipoxygenase, phosphoinositide 3-kinase, sphingosine kinase and sphingomyelinase. These enzymes and their downstream targets constitute a complex lipid signaling network with multiple nodes of interaction and cross-regulation. Imbalances in this network contribute to the pathogenesis of human disease. Although the function of a particular signaling lipid is traditionally studied in isolation, this review attempts a more integrated overview of the key role of these signaling lipids in inflammation, cancer and metabolic disease, and discusses emerging strategies for therapeutic intervention.
- 2Yang, H.; Rudge, D. G.; Koos, J. D.; Vaidialingam, B.; Yang, H. J.; Pavletich, N. P. mTOR kinase structure, mechanism and regulation. Nature 2013, 497 (7448), 217– 223, DOI: 10.1038/nature12122Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvFyns7k%253D&md5=07809555fbdf2004819a7f2da5727bafmTOR kinase structure, mechanism and regulationYang, Haijuan; Rudge, Derek G.; Koos, Joseph D.; Vaidialingam, Bhamini; Yang, Hyo J.; Pavletich, Nikola P.Nature (London, United Kingdom) (2013), 497 (7448), 217-223CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)The mammalian target of rapamycin (mTOR), a phosphoinositide 3-kinase-related protein kinase, controls cell growth in response to nutrients and growth factors and is frequently deregulated in cancer. Here we report co-crystal structures of a complex of truncated mTOR and mammalian lethal with SEC13 protein 8 (mLST8) with an ATP transition state mimic (MgF3-) and with ATP-site inhibitors (Torin2, PP242, and PI-103). The structures reveal an intrinsically active kinase conformation, with catalytic residues and a catalytic mechanism remarkably similar to canonical protein kinases. The active site is highly recessed owing to the FKBP12-rapamycin-binding (FRB) domain and an inhibitory helix protruding from the catalytic cleft. MTOR-activating mutations map to the structural framework that holds these elements in place, indicating that the kinase is controlled by restricted access. In vitro biochem. shows that the FRB domain acts as a gatekeeper, with its rapamycin-binding site interacting with substrates to grant them access to the restricted active site. Rapamycin-FKBP12 inhibits the kinase by directly blocking substrate recruitment and by further restricting active-site access. The structures also reveal active-site residues and conformational changes that underlie inhibitor potency and specificity.
- 3Sarbassov, D. D.; Guertin, D. A.; Ali, S. M.; Sabatini, D. M. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005, 307 (5712), 1098– 1101, DOI: 10.1126/science.1106148Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXhtlSrtbY%253D&md5=de758fd8128561e34301e67a42ae13a6Phosphorylation and Regulation of Akt/PKB by the Rictor-mTOR ComplexSarbassov, Dos D.; Guertin, David A.; Ali, Siraj M.; Sabatini, David M.Science (Washington, DC, United States) (2005), 307 (5712), 1098-1101CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Deregulation of Akt/protein kinase B (PKB) is implicated in the pathogenesis of cancer and diabetes. Akt/PKB activation requires the phosphorylation of Thr308 in the activation loop by the phosphoinositide-dependent kinase 1 (PDK1) and Ser473 within the carboxyl-terminal hydrophobic motif by an unknown kinase. We show that in Drosophila and human cells the target of rapamycin (TOR) kinase and its assocd. protein rictor are necessary for Ser473 phosphorylation and that a redn. in rictor or mammalian TOR (mTOR) expression inhibited an Akt/PKB effector. The rictor-mTOR complex directly phosphorylated Akt/PKB on Ser473 in vitro and facilitated Thr308 phosphorylation by PDK1. Rictor-mTOR may serve as a drug target in tumors that have lost the expression of PTEN, a tumor suppressor that opposes Akt/PKB activation.
- 4Feng, J.; Park, J.; Cron, P.; Hess, D.; Hemmings, B. A. Identification of a PKB/Akt hydrophobic motif Ser-473 kinase as DNA-dependent protein kinase. J. Biol. Chem. 2004, 279 (39), 41189– 41196, DOI: 10.1074/jbc.M406731200Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXnslWms70%253D&md5=b6bad6828d91d47e2b36f537ca6d5f85Identification of a PKB/Akt Hydrophobic Motif Ser-473 Kinase as DNA-dependent Protein KinaseFeng, Jianhua; Park, Jongsun; Cron, Peter; Hess, Daniel; Hemmings, Brian A.Journal of Biological Chemistry (2004), 279 (39), 41189-41196CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Full activation of protein kinase B (PKB)/Akt requires phosphorylation on Thr-308 and Ser-473 by 3-phosphoinositide-dependent kinase-1 (PDK1) and Ser-473 kinase (S473K), resp. Although PDK1 has been well characterized, the identification of the S473K remains controversial. A major PKB Ser-473 kinase activity was purified from the membrane fraction of HEK293 cells and found to be DNA-dependent protein kinase (DNA-PK). DNA-PK co-localized and assocd. with PKB at the plasma membrane. In vitro, DNA-PK phosphorylated PKB on Ser-473, resulting in a ∼10-fold enhancement of PKB activity. Knockdown of DNA-PK by small interfering RNA inhibited Ser-473 phosphorylation induced by insulin and pervanadate. DNA-PK-deficient glioblastoma cells did not respond to insulin at the level of Ser-473 phosphorylation; this effect was restored by complementation with the human PRKDC gene. We conclude that DNA-PK is a long sought after kinase responsible for the Ser-473 phosphorylation step in the activation of PKB.
- 5Wymann, M. P.; Marone, R. Phosphoinositide 3-kinase in disease: timing, location, and scaffolding. Curr. Opin. Cell Biol. 2005, 17 (2), 141– 149, DOI: 10.1016/j.ceb.2005.02.011Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXisVCqtb4%253D&md5=d126dfae131b86e73e8842ec9149fd01Phosphoinositide 3-kinase in disease: timing, location, and scaffoldingWymann, Matthias P.; Marone, RominaCurrent Opinion in Cell Biology (2005), 17 (2), 141-149CODEN: COCBE3; ISSN:0955-0674. (Elsevier Ltd.)A review. When PI3Ks are deregulated by aberrant surface receptors or modulators, accumulation of PtdIns(3,4,5)P3 leads to increased cell growth, proliferation and contact-independent survival. The PI3K/PKB/TOR axis controls protein synthesis and growth, while PtdIns(3,4,5)P3-mediated activation of Rho GTPases directs cell motility. PI3K activity has been linked to the formation of tumors, metastasis, chronic inflammation, allergy and cardiovascular disease. Although increased PtdIns(3,4,5)P3 is a well-established cause of disease, it is seldom known which PI3K isoform is implied. Recent work has demonstrated that PI3Kγ contributes to the control of cAMP levels in the cardiac system, where the protein acts as a scaffold, but not as a lipid kinase.
- 6Bozulic, L.; Hemmings, B. A. PIKKing on PKB: regulation of PKB activity by phosphorylation. Curr. Opin. Cell Biol. 2009, 21 (2), 256– 261, DOI: 10.1016/j.ceb.2009.02.002Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXksVClu7Y%253D&md5=0b7a3e6ea561f8479701a01133b5d30cPIKKing on PKB: regulation of PKB activity by phosphorylationBozulic, Lana; Hemmings, Brian A.Current Opinion in Cell Biology (2009), 21 (2), 256-261CODEN: COCBE3; ISSN:0955-0674. (Elsevier B.V.)A review. Protein kinase B (PKB)/Akt kinase is a key regulator of a wide range of cellular processes including growth, proliferation and survival. PKB is clearly a crucial signaling mol. and extensive research efforts aim to understand its regulation and action. Recent studies of the regulation of PKB activity by hydrophobic motif phosphorylation have yielded several exciting findings about members of the phosphatidylinositol 3-kinase (PI3K)-like family of kinases (PIKKs) acting as PKB regulators. Mammalian target of rapamycin complex 2 (mTORC2) and DNA-dependent protein kinase (DNA-PK) can both phosphorylate Ser-473 and activate PKB. This review concerns PKB regulation by mTORC2 and DNA-PK in a stimulus-dependent and context-dependent manner and the possible implications of this for PKB activity, substrate specificity, and therapeutic intervention.
- 7Magnuson, B.; Ekim, B.; Fingar, D. C. Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem. J. 2012, 441 (1), 1– 21, DOI: 10.1042/BJ20110892Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs1Cgs7jK&md5=5d69e3be00deef02d3fce1ed2716873dRegulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networksMagnuson, Brian; Ekim, Bilgen; Fingar, Diane C.Biochemical Journal (2012), 441 (1), 1-21CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)A review. The ribosomal protein S6K (S6 kinase) represents an extensively studied effector of the TORC1 [TOR (target of rapamycin) complex 1], which possesses important yet incompletely defined roles in cellular and organismal physiol. TORC1 functions as an environmental sensor by integrating signals derived from diverse environmental cues to promote anabolic and inhibit catabolic cellular functions. mTORC1 (mammalian TORC1) phosphorylates and activates S6K1 and S6K2, whose first identified substrate was rpS6 (ribosomal protein S6), a component of the 40S ribosome. Studies over the past decade have uncovered a no. of addnl. S6K1 substrates, revealing multiple levels at which the mTORC1-S6K1 axis regulates cell physiol. The results thus far indicate that the mTORC1-S6K1 axis controls fundamental cellular processes, including transcription, translation, protein and lipid synthesis, cell growth/size and cell metab. In the present review we summarize the regulation of S6Ks, their cellular substrates and functions, and their integration within rapidly expanding mTOR (mammalian TOR) signalling networks. Although our understanding of the role of mTORC1-S6K1 signalling in physiol. remains in its infancy, evidence indicates that this signalling axis controls, at least in part, glucose homeostasis, insulin sensitivity, adipocyte metab., body mass and energy balance, tissue and organ size, learning, memory and aging. As dysregulation of this signalling axis contributes to diverse disease states, improved understanding of S6K regulation and function within mTOR signalling networks may enable the development of novel therapeutics.
- 8Laplante, M.; Sabatini, D. M. mTOR signaling in growth control and disease. Cell 2012, 149 (2), 274– 293, DOI: 10.1016/j.cell.2012.03.017Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xls1eguro%253D&md5=d1adac8ec64da63358e0af26a17ceb4emTOR signaling in growth control and diseaseLaplante, Mathieu; Sabatini, David M.Cell (Cambridge, MA, United States) (2012), 149 (2), 274-293CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The mechanistic target of rapamycin (mTOR) signaling pathway senses and integrates a variety of environmental cues to regulate organismal growth and homeostasis. The pathway regulates many major cellular processes and is implicated in an increasing no. of pathol. conditions, including cancer, obesity, type 2 diabetes, and neurodegeneration. Here, we review recent advances in our understanding of the mTOR pathway and its role in health, disease, and aging. We further discuss pharmacol. approaches to treat human pathologies linked to mTOR deregulation.
- 9Saxton, R. A.; Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 2017, 168 (6), 960– 976, DOI: 10.1016/j.cell.2017.02.004Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkt1Ogtb4%253D&md5=3fdee3d04bf88d3aafc532d4e2f1e2dcmTOR signaling in growth, metabolism, and diseaseSaxton, Robert A.; Sabatini, David M.Cell (Cambridge, MA, United States) (2017), 168 (6), 960-976CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. The mechanistic target of rapamycin (mTOR) coordinates eukaryotic cell growth and metab. with environmental inputs, including nutrients and growth factors. Extensive research over the past two decades has established a central role for mTOR in regulating many fundamental cell processes, from protein synthesis to autophagy, and deregulated mTOR signaling is implicated in the progression of cancer and diabetes, as well as the aging process. Here, we review recent advances in our understanding of mTOR function, regulation, and importance in mammalian physiol. We also highlight how the mTOR signaling network contributes to human disease and discuss the current and future prospects for therapeutically targeting mTOR in the clinic.
- 10Vivanco, I.; Sawyers, C. L. The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat. Rev. Cancer 2002, 2 (7), 489– 501, DOI: 10.1038/nrc839Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XkvFKltLs%253D&md5=c6762d32f9f1281632f4ccdbcd29268cThe phosphatidylinositol 3-Kinase-AKT pathway in human cancerVivanco, Igor; Sawyers, Charles L.Nature Reviews Cancer (2002), 2 (7), 489-501CODEN: NRCAC4; ISSN:1474-175X. (Nature Publishing Group)A review. One signal that is overactivated in a wide range of tumor types is the prodn. of a phospholipid, phosphatidylinositol (3,4,5) trisphosphate, by phosphatidylinositol 3-kinase (PI3K). This lipid and the protein kinase that is activated by it, AKT, trigger a cascade of responses, from cell growth and proliferation to survival and motility, that drive tumor progression. Small-mol. therapeutics that block PI3K signaling might deal a severe blow to cancer cells by blocking many aspects of the tumor-cell phenotype.
- 11Marone, R.; Cmiljanovic, V.; Giese, B.; Wymann, M. P. Targeting phosphoinositide 3-kinase: moving towards therapy. Biochim. Biophys. Acta, Proteins Proteomics 2008, 1784 (1), 159– 185, DOI: 10.1016/j.bbapap.2007.10.003Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXptlerug%253D%253D&md5=bf9f259b3da61067631823656658e0f7Targeting phosphoinositide 3-kinase-Moving towards therapyMarone, Romina; Cmiljanovic, Vladimir; Giese, Bernd; Wymann, Matthias P.Biochimica et Biophysica Acta, Proteins and Proteomics (2008), 1784 (1), 159-185CODEN: BBAPBW; ISSN:1570-9639. (Elsevier Ltd.)A review. Phosphoinositide 3-kinases (PI3K) orchestrate cell responses including mitogenic signaling, cell survival and growth, metabolic control, vesicular trafficking, degranulation, cytoskeletal rearrangement and migration. Deregulation of the PI3K pathway occurs by activating mutations in growth factor receptors or the PIK3CA locus coding for PI3Kα, by loss of function of the lipid phosphatase and tensin homolog deleted in chromosome ten (PTEN/MMAC/TEP1), by the up-regulation of protein kinase B (PKB/Akt), or the impairment of the tuberous sclerosis complex (TSC1/2). All these events are linked to growth and proliferation, and have thus prompted a significant interest in the pharmaceutical targeting of the PI3K pathway in cancer. Genetic targeting of PI3Kγ (p110γ) and PI3Kδ (p110δ) in mice has underlined a central role of these PI3K isoforms in inflammation and allergy, as they modulate chemotaxis of leukocytes and degranulation in mast cells. Proof-of-concept mols. selective for PI3Kγ have already successfully alleviated disease progress in murine models of rheumatoid arthritis and lupus erythematosus. As targeting PI3K moves forward to therapy of chronic, non-fatal disease, safety concerns for PI3K inhibitors increase. Many of the present inhibitor series interfere with target of rapamycin (TOR), DNA-dependent protein kinase (DNA-PKcs) and activity of the ataxia telangiectasia mutated gene product (ATM). Here we review the current disease-relevant knowledge for isoform-specific PI3K function in the above mentioned diseases, and review the progress of > 400 recent patents covering pharmaceutical targeting of PI3K. Currently, several drugs targeting the PI3K pathway have entered clin. trials (phase I) for solid tumors and suppression of tissue damage after myocardial infarction (phases I,II).
- 12Wymann, M. PI3Ks—Drug Targets in Inflammation and Cancer. In Phosphoinositides I: Enzymes of Synthesis and Degradation; Balla, T., Wymann, M., York, J. D., Eds.; Springer: Dordrecht, The Netherlands, 2012; pp 111– 181.Google ScholarThere is no corresponding record for this reference.
- 13Choi, J.; Chen, J.; Schreiber, S. L.; Clardy, J. Structure of the FKBP1 2-rapamycin complex interacting with the binding domain of human FRAP. Science 1996, 273 (5272), 239– 242, DOI: 10.1126/science.273.5272.239Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK28Xkt1Crs7k%253D&md5=c72a82e14c09d9a705215b4ac0525336Structure of the FKBP12-rapamycin complex interacting with the binding domain of human FRAPChoi, Jungwon; Chen, Jie; Schreiber, Stuart L.; Clardy, JonScience (Washington, D. C.) (1996), 273 (5272), 239-242CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Rapamycin, a potent immunosuppressive agent, binds two proteins: the FK506-binding protein (FKBP12) and the FKBP-rapamycin-assocd. protein (FRAP). A crystal structure of the ternary complex of human FKBP12, rapamycin, and the FKBP12-rapamycin-binding (FRB) domain of human FRAP at a resoln. of 2.7 angstroms revealed the two proteins bound together as a result of the ability of rapamycin to occupy two different hydrophobic binding pockets simultaneously. The structure shows extensive interactions between rapamycin and both proteins, but fewer interactions between the proteins. The structure of the FRB domain of FRAP clarifies both rapamycin-independent and -dependent effects obsd. for mutants of FRAP and its homologs in the family of proteins related to the ataxia-telangiectasis mutant gene product, and it illustrates how a small cell-permeable mol. can mediate protein dimerization.
- 14Hudes, G.; Carducci, M.; Tomczak, P.; Dutcher, J.; Figlin, R.; Kapoor, A.; Staroslawska, E.; Sosman, J.; McDermott, D.; Bodrogi, I.; Kovacevic, Z.; Lesovoy, V.; Schmidt-Wolf, I. G. H.; Barbarash, O.; Gokmen, E.; O’Toole, T.; Lustgarten, S.; Moore, L.; Motzer, R. J. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 2007, 356 (22), 2271– 2281, DOI: 10.1056/NEJMoa066838Google Scholar14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXmtVKkurs%253D&md5=2fc7685df067fd6ea813483610e89dc8Temsirolimus, interferon alfa, or both for advanced renal-cell carcinomaHudes, Gary; Carducci, Michael; Tomczak, Piotr; Dutcher, Janice; Figlin, Robert; Kapoor, Anil; Staroslawska, Elzbieta; Sosman, Jeffrey; McDermott, David; Bodrogi, Istvan; Kovacevic, Zoran; Lesovoy, Vladimir; Schmidt-Wolf, Ingo G. H.; Barbarash, Olga; Gokmen, Erhan; O'Toole, Timothy; Lustgarten, Stephanie; Moore, Laurence; Motzer, Robert J.New England Journal of Medicine (2007), 356 (22), 2271-2281CODEN: NEJMAG; ISSN:0028-4793. (Massachusetts Medical Society)Interferon alfa is widely used for metastatic renal-cell carcinoma but has limited efficacy and tolerability. Temsirolimus, a specific inhibitor of the mammalian target of rapamycin kinase, may benefit patients with this disease. In this multicenter, phase 3 trial, we randomly assigned 626 patients with previously untreated, poor-prognosis metastatic renal-cell carcinoma to receive 25 mg of i.v. temsirolimus weekly, 3 million U of interferon alfa (with an increase to 18 million U) s.c. three times weekly, or combination therapy with 15 mg of temsirolimus weekly plus 6 million U of interferon alfa three times weekly. The primary end point was overall survival in comparisons of the temsirolimus group and the combination-therapy group with the interferon group. RESULTS Patients who received temsirolimus alone had longer overall survival (hazard ratio for death, 0.73; 95% confidence interval [CI], 0.58 to 0.92; P = 0.008) and progression-free survival (P < 0.001) than did patients who received interferon alone. Overall survival in the combination-therapy group did not differ significantly from that in the interferon group (hazard ratio, 0.96; 95% CI, 0.76 to 1.20; P = 0.70). Median overall survival times in the interferon group, the temsirolimus group, and the combination-therapy group were 7.3, 10.9, and 8.4 mo, resp. Rash, peripheral edema, hyperglycemia, and hyperlipidemia were more common in the temsirolimus group, whereas asthenia was more common in the interferon group. There were fewer patients with serious adverse events in the temsirolimus group than in the interferon group (P = 0.02). As compared with interferon alfa, temsirolimus improved overall survival among patients with metastatic renal-cell carcinoma and a poor prognosis. The addn. of temsirolimus to interferon did not improve survival. (ClinicalTrials.gov no., NCT00065468.).
- 15Motzer, R. J.; Escudier, B.; Oudard, S.; Hutson, T. E.; Porta, C.; Bracarda, S.; Grünwald, V.; Thompson, J. A.; Figlin, R. A.; Hollaender, N.; Urbanowitz, G.; Berg, W. J.; Kay, A.; Lebwohl, D.; Ravaud, A. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008, 372 (9637), 449– 456, DOI: 10.1016/S0140-6736(08)61039-9Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXps1GmsLY%253D&md5=cbfa5d207cfab693b3bccd24963380f0Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomized, placebo-controlled phase III trialMotzer, Robert J.; Escudier, Bernard; Oudard, Stephane; Hutson, Thomas E.; Porta, Camillo; Bracarda, Sergio; Gruenwald, Viktor; Thompson, John A.; Figlin, Robert A.; Hollaender, Norbert; Urbanowitz, Gladys; Berg, William J.; Kay, Andrea; Lebwohl, David; Ravaud, AlainLancet (2008), 372 (9637), 449-456CODEN: LANCAO; ISSN:0140-6736. (Elsevier Ltd.)Everolimus (RAD001) is an orally administered inhibitor of the mammalian target of rapamycin (mTOR), a therapeutic target for metastatic renal cell carcinoma. We did a phase III, randomized, double-blind, placebo-controlled trial of everolimus in patients with metastatic renal cell carcinoma whose disease had progressed on vascular endothelial growth factor-targeted therapy. Patients with metastatic renal cell carcinoma which had progressed on sunitinib, sorafenib, or both, were randomly assigned in a two to one ratio to receive everolimus 10 mg once daily (n=272) or placebo (n=138), in conjunction with best supportive care. Randomisation was done centrally via an interactive voice response system using a validated computer system, and was stratified by Memorial Sloan-Kettering Cancer Center prognostic score and previous anticancer therapy, with a permuted block size of six. The primary endpoint was progression-free survival, assessed via a blinded, independent central review. The study was designed to be terminated after 290 events of progression. Anal. was by intention to treat. This study is registered with, no. All randomized patients were included in efficacy analyses. The results of the second interim anal. indicated a significant difference in efficacy between arms and the trial was thus halted early after 191 progression events had been obsd. (101 [37%] events in the everolimus group, 90 [65%] in the placebo group; hazard ratio 0.30, 95% CI 0.22-0.40, p<0.0001; median progression-free survival 4.0 [95% CI 3.7-5.5] vs 1.9 [1.8-1.9] months). Stomatitis (107 [40%] patients in the everolimus group vs 11 [8%] in the placebo group), rash (66 [25%] vs six [4%]), and fatigue (53 [20%] vs 22 [16%]) were the most commonly reported adverse events, but were mostly mild or moderate in severity. Pneumonitis (any grade) was detected in 22 (8%) patients in the everolimus group, of whom eight had pneumonitis of grade 3 severity. Treatment with everolimus prolongs progression-free survival relative to placebo in patients with metastatic renal cell carcinoma that had progressed on other targeted therapies. Novartis Oncol.
- 16Jerusalem, G.; Rorive, A.; Collignon, J. Use of mTOR inhibitors in the treatment of breast cancer: an evaluation of factors that influence patient outcomes. Breast Cancer: Targets Ther. 2014, 6, 43– 57, DOI: 10.2147/BCTT.S38679Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs12gsrjE&md5=9a12c70c7c917baa41d282bc135b12afUse of mTOR inhibitors in the treatment of breast cancer: an evaluation of factors that influence patient outcomesJerusalem, Guy; Rorive, Andree; Collignon, JoelleBreast Cancer: Targets and Therapy (2014), 6 (), 43-57, 15CODEN: BCTTA9; ISSN:1179-1314. (Dove Medical Press Ltd.)A review. Many systemic treatment options are available for advanced breast cancer, including endocrine therapy, chemotherapy, anti-human epidermal growth factor receptor 2 (HER2) therapy, and other targeted agents. Recently, everolimus, a mammalian target of rapamycin (mTOR) inhibitor, combined with exemestane, an aromatase inhibitor, has been approved in Europe and the USA for patients suffering from estrogen receptor-pos., HER2-neg. advanced breast cancer previously treated by a nonsteroidal aromatase inhibitor, based on the results of BOLERO-2 (Breast cancer trials of OraL EveROlimus). This study showed a statistically significant and clin. meaningful improvement in median progression-free survival. Results concerning the impact on overall survival are expected in the near future. This clin. oriented review focuses on the use of mTOR inhibitors in breast cancer. Results reported with first-generation mTOR inhibitors (ridaforolimus, temsirolimus, everolimus) are discussed. The current and potential role of mTOR inhibitors is reported according to breast cancer subtype (estrogen receptor-pos. HER2-neg., triple-neg., and HER2-pos. ER-pos./neg. disease). Everolimus is currently being evaluated in the adjuvant setting in high-risk estrogen receptor-pos., HER2-neg. early breast cancer. Continuing mTOR inhibition or alternatively administering other drugs targeting the phosphatidylinositol-3-kinase/protein kinase B-mTOR pathway after progression on treatments including an mTOR inhibitor is under evaluation. Potential biomarkers to select patients showing a more pronounced benefit are reviewed, but we are not currently using these biomarkers in routine practice. Subgroup anal. of BOLERO 2 has shown that the benefit is consistent in all subgroups and that it is impossible to select patients not benefiting from addn. of everolimus to exemestane. Side effects and impact on quality of life are other important issues discussed in this review. Second-generation mTOR inhibitors and dual mTOR-phosphatidylinositol-3-kinase inhibitors are currently being evaluated in clin. trials.
- 17André, F.; O’Regan, R.; Ozguroglu, M.; Toi, M.; Xu, B.; Jerusalem, G.; Masuda, N.; Wilks, S.; Arena, F.; Isaacs, C.; Yap, Y.-S.; Papai, Z.; Lang, I.; Armstrong, A.; Lerzo, G.; White, M.; Shen, K.; Litton, J.; Chen, D.; Zhang, Y.; Ali, S.; Taran, T.; Gianni, L. Everolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trial. Lancet Oncol. 2014, 15 (6), 580– 591, DOI: 10.1016/S1470-2045(14)70138-XGoogle Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmtlWnsr4%253D&md5=cfbb16368227cc80463ef26c2593b69dEverolimus for women with trastuzumab-resistant, HER2-positive, advanced breast cancer (BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 trialAndre, Fabrice; O'Regan, Ruth; Ozguroglu, Mustafa; Toi, Masakazu; Xu, Binghe; Jerusalem, Guy; Masuda, Norikazu; Wilks, Sharon; Arena, Francis; Isaacs, Claudine; Yap, Yoon-Sim; Papai, Zsuzsanna; Lang, Istvan; Armstrong, Anne; Lerzo, Guillermo; White, Michelle; Shen, Kunwei; Litton, Jennifer; Chen, David; Zhang, Yufen; Ali, Shyanne; Taran, Tetiana; Gianni, LucaLancet Oncology (2014), 15 (6), 580-591CODEN: LOANBN; ISSN:1470-2045. (Elsevier Ltd.)Disease progression in patients with HER2-pos. breast cancer receiving trastuzumab might be assocd. with activation of the PI3K/Akt/mTOR intracellular signalling pathway. We aimed to assess whether the addn. of the mTOR inhibitor everolimus to trastuzumab might restore sensitivity to trastuzumab. In this randomised, double-blind, placebo-controlled, phase 3 trial, we recruited women with HER2-pos., trastuzumab-resistant, advanced breast carcinoma who had previously received taxane therapy. Eligible patients were randomly assigned (1:1) using a central patient screening and randomization system to daily everolimus (5 mg/day) plus weekly trastuzumab (2 mg/kg) and vinorelbine (25 mg/m2) or to placebo plus trastuzumab plus vinorelbine, in 3-wk cycles, stratified by previous lapatinib use. The primary endpoint was progression-free survival (PFS) by local assessment in the intention-to-treat population. We report the final anal. for PFS; overall survival follow-up is still in progress. This trial is registered with ClinicalTrials.gov, no. NCT01007942.Between Oct 26, 2009, and May 23, 2012, 569 patients were randomly assigned to everolimus (n=284) or placebo (n=285). Median follow-up at the time of anal. was 20·2 mo (IQR 15·0-27·1). Median PFS was 7·00 mo (95% CI 6·74-8·18) with everolimus and 5·78 mo (5·49-6·90) with placebo (hazard ratio 0·78 [95% CI 0·65-0·95]; p=0·0067). The most common grade 3-4 adverse events were neutropenia (204 [73%] of 280 patients in the everolimus group vs 175 [62%] of 282 patients in the placebo group), leucopenia (106 [38%] vs 82 [29%]), anemia (53 [19%] vs 17 [6%]), febrile neutropenia (44 [16%] vs ten [4%]), stomatitis (37 [13%] vs four [1%]), and fatigue (34 [12%] vs 11 [4%]). Serious adverse events were reported in 117 (42%) patients in the everolimus group and 55 (20%) in the placebo group; two on-treatment deaths due to adverse events occurred in each group. The addn. of everolimus to trastuzumab plus vinorelbine significantly prolongs PFS in patients with trastuzumab-resistant and taxane-pretreated, HER2-pos., advanced breast cancer. The clin. benefit should be considered in the context of the adverse event profile in this population. Novartis Pharmaceuticals Corporation.
- 18Peterson, M. E. Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trial. Supportive care in cancer: official journal of the Multinational Association of Supportive Care in Cancer 2013, 21 (8), 2341– 2349, DOI: 10.1007/s00520-013-1826-3Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3snkvVKksw%253D%253D&md5=21bfc1094be58365540d6fe494138198Management of adverse events in patients with hormone receptor-positive breast cancer treated with everolimus: observations from a phase III clinical trialPeterson Mary ESupportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer (2013), 21 (8), 2341-9 ISSN:.Everolimus is a mammalian target of rapamycin (mTOR) inhibitor approved for the treatment of advanced renal cell carcinoma, pancreatic neuroendocrine tumors, subependymal giant cell astrocytoma associated with tuberous sclerosis complex, renal angiomyolipoma and tuberous sclerosis complex, and, in combination with exemestane, for hormone receptor-positive HER2-negative advanced breast cancer after failure of treatment with letrozole or anastrozole. Results from the phase III BOLERO-2 trial demonstrated that everolimus in combination with exemestane provided significant clinical benefit to patients with advanced hormone receptor-positive breast cancer. Although everolimus is generally well tolerated, as with most therapies administered in an advanced cancer setting, drug-related adverse events (AEs) inevitably occur. Most common AEs observed in the everolimus studies include stomatitis, rash, infection, noninfectious pneumonitis, and hyperglycemia. Clinical awareness and early identification of such AEs by oncology nurses are essential to dosing (interruptions, reduction, and treatment discontinuation); quality of life; and, ultimately, patient outcomes. Because everolimus has already been shown to significantly improve clinical efficacy in patients with advanced breast cancer, a proactive approach to the practical management of AEs associated with this mTOR inhibitor as well as other most common AEs observed in this patient population has been reviewed and outlined here.
- 19Santulli, G.; Totary-Jain, H. Tailoring mTOR-based therapy: molecular evidence and clinical challenges. Pharmacogenomics 2013, 14 (12), 1517– 1526, DOI: 10.2217/pgs.13.143Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhsVWrurrN&md5=0a3798fb659843d231910151b2f148edTailoring mTOR-based therapy: molecular evidence and clinical challengesSantulli, Gaetano; Totary-Jain, HanaPharmacogenomics (2013), 14 (12), 1517-1526CODEN: PARMFL; ISSN:1462-2416. (Future Medicine Ltd.)A review. The mTOR signaling pathway integrates inputs from a variety of upstream stimuli to regulate diverse cellular processes including proliferation, growth, survival, motility, autophagy, protein synthesis and metab. The mTOR pathway is dysregulated in a no. of human pathologies including cancer, diabetes, obesity, autoimmune disorders, neurol. disease and aging. Ongoing clin. trials testing mTOR-targeted treatments no. in the hundreds and underscore its therapeutic potential. To date mTOR inhibitors are clin. approved to prevent organ rejection, to inhibit restenosis after angioplasty, and to treat several advanced cancers. In this review we discuss the continuously evolving field of mTOR pharmacogenomics, as well as highlight the emerging efforts in identifying diagnostic and prognostic markers, including miRNAs, in order to assess successful therapeutic responses.
- 20Meng, L. H.; Zheng, X. F. Toward rapamycin analog (rapalog)-based precision cancer therapy. Acta Pharmacol. Sin. 2015, 36 (10), 1163– 1169, DOI: 10.1038/aps.2015.68Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhs1Wls7jP&md5=af1357a49b132c2264ca74577ae2ae48Toward rapamycin analog (rapalog)-based precision cancer therapyMeng, Ling-hua; Zheng, X. F. StevenActa Pharmacologica Sinica (2015), 36 (10), 1163-1169CODEN: APSCG5; ISSN:1671-4083. (Nature Publishing Group)Rapamycin and its analogs (rapalogs) are the first generation of mTOR inhibitors, which have the same mol. scaffold, but different physiochem. properties. Rapalogs are being tested in a wide spectrum of human tumors as both monotherapy and a component of combination therapy. Among them, temsirolimus and everolimus have been approved for the treatment of breast and renal cancer. However, objective response rates with rapalogs in clin. trials are modest and variable. Identification of biomarkers predicting response to rapalogs, and discovery of drug combinations with improved efficacy and tolerated toxicity are crit. to moving this class of targeted therapeutics forward. This review focuses on the aberrations in the PI3K/mTOR pathway in human tumor cells or tissues as predictive biomarkers for rapalog efficacy. Recent results of combinational therapy using rapalogs and other anticancer drugs are documented. With the rapid development of next-generation genomic sequencing and precision medicine, rapalogs will provide greater benefits to cancer patients.
- 21Feldman, M. E.; Apsel, B.; Uotila, A.; Loewith, R.; Knight, Z. A.; Ruggero, D.; Shokat, K. M. Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol. 2009, 7 (2), e1000038, DOI: 10.1371/journal.pbio.1000038Google ScholarThere is no corresponding record for this reference.
- 22Kang, S. A.; Pacold, M. E.; Cervantes, C. L.; Lim, D.; Lou, H. J.; Ottina, K.; Gray, N. S.; Turk, B. E.; Yaffe, M. B.; Sabatini, D. M. mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin. Science 2013, 341 (6144), 1236566, DOI: 10.1126/science.1236566Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3sfktVWmsw%253D%253D&md5=904daf44e853714de3fb540a359e2546mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycinKang Seong A; Pacold Michael E; Cervantes Christopher L; Lim Daniel; Lou Hua Jane; Ottina Kathleen; Gray Nathanael S; Turk Benjamin E; Yaffe Michael B; Sabatini David MScience (New York, N.Y.) (2013), 341 (6144), 1236566 ISSN:.The mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) protein kinase promotes growth and is the target of rapamycin, a clinically useful drug that also prolongs life span in model organisms. A persistent mystery is why the phosphorylation of many bona fide mTORC1 substrates is resistant to rapamycin. We find that the in vitro kinase activity of mTORC1 toward peptides encompassing established phosphorylation sites varies widely and correlates strongly with the resistance of the sites to rapamycin, as well as to nutrient and growth factor starvation within cells. Slight modifications of the sites were sufficient to alter mTORC1 activity toward them in vitro and to cause concomitant changes within cells in their sensitivity to rapamycin and starvation. Thus, the intrinsic capacity of a phosphorylation site to serve as an mTORC1 substrate, a property we call substrate quality, is a major determinant of its sensitivity to modulators of the pathway. Our results reveal a mechanism through which mTORC1 effectors can respond differentially to the same signals.
- 23Jacinto, E.; Loewith, R.; Schmidt, A.; Lin, S.; Ruegg, M. A.; Hall, A.; Hall, M. N. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat. Cell Biol. 2004, 6 (11), 1122– 8, DOI: 10.1038/ncb1183Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptFentL8%253D&md5=42d53ee5d9dfc24287cf05fd29f41aa8Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitiveJacinto, Estela; Loewith, Robbie; Schmidt, Anja; Lin, Shuo; Rueegg, Markus A.; Hall, Alan; Hall, Michael N.Nature Cell Biology (2004), 6 (11), 1122-1128CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)The target of rapamycin (TOR) is a highly conserved protein kinase and a central controller of cell growth. In budding yeast, TOR is found in structurally and functionally distinct protein complexes: TORC1 and TORC2. A mammalian counterpart of TORC1 (mTORC1) has been described, but it is not known whether TORC2 is conserved in mammals. Here, the authors report that a mammalian counterpart of TORC2 (mTORC2) also exists. The mTORC2 contains mTOR, mLST8 and mAVO3, but not raptor. Like yeast TORC2, mTORC2 is rapamycin insensitive and seems to function upstream of Rho GTPases to regulate the actin cytoskeleton. The mTORC2 is not upstream of the mTORC1 effector S6K. Thus, two distinct TOR complexes constitute a primordial signaling network conserved in eukaryotic evolution to control the fundamental process of cell growth.
- 24O’Reilly, K. E.; Rojo, F.; She, Q. B.; Solit, D.; Mills, G. B.; Smith, D.; Lane, H.; Hofmann, F.; Hicklin, D. J.; Ludwig, D. L.; Baselga, J.; Rosen, N. mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006, 66 (3), 1500– 1508, DOI: 10.1158/0008-5472.CAN-05-2925Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsVegtg%253D%253D&md5=758cd9999dc156bf498edf32174fc2e7mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates AktO'Reilly, Kathryn E.; Rojo, Fredi; She, Qing-Bai; Solit, David; Mills, Gordon B.; Smith, Debra; Lane, Heidi; Hofmann, Francesco; Hicklin, Daniel J.; Ludwig, Dale L.; Baselga, Jose; Rosen, NealCancer Research (2006), 66 (3), 1500-1508CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Stimulation of the insulin and insulin-like growth factor I (IGF-I) receptor activates the phosphoinositide-3-kinase/Akt/mTOR pathway causing pleiotropic cellular effects including an mTOR-dependent loss in insulin receptor substrate-1 expression leading to feedback down-regulation of signaling through the pathway. In model systems, tumors exhibiting mutational activation of phosphoinositide-3-kinase/Akt kinase, a common event in cancers, are hypersensitive to mTOR inhibitors, including rapamycin. Despite the activity in model systems, in patients, mTOR inhibitors exhibit more modest antitumor activity. We now show that mTOR inhibition induces insulin receptor substrate-1 expression and abrogates feedback inhibition of the pathway, resulting in Akt activation both in cancer cell lines and in patient tumors treated with the rapamycin deriv., RAD001. IGF-I receptor inhibition prevents rapamycin-induced Akt activation and sensitizes tumor cells to inhibition of mTOR. In contrast, IGF-I reverses the antiproliferative effects of rapamycin in serum-free medium. The data suggest that feedback down-regulation of receptor tyrosine kinase signaling is a frequent event in tumor cells with constitutive mTOR activation. Reversal of this feedback loop by rapamycin may attenuate its therapeutic effects, whereas combination therapy that ablates mTOR function and prevents Akt activation may have improved antitumor activity.
- 25Liu, P.; Gan, W.; Chin, Y. R.; Ogura, K.; Guo, J.; Zhang, J.; Wang, B.; Blenis, J.; Cantley, L. C.; Toker, A.; Su, B.; Wei, W. PtdIns(3,4,5)P3-dependent activation of the mTORC2 kinase complex. Cancer Discovery 2015, 5 (11), 1194– 1209, DOI: 10.1158/2159-8290.CD-15-0460Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvVWqsr3E&md5=a218e7e37d78aaa532a8ff44db230447PtdIns(3,4,5)P3-dependent activation of the mtorc2 kinase complexLiu, Pengda; Gan, Wenjian; Chin, Y. Rebecca; Ogura, Kohei; Guo, Jianping; Zhang, Jinfang; Wang, Bin; Blenis, John; Cantley, Lewis C.; Toker, Alex; Su, Bing; Wei, WenyiCancer Discovery (2015), 5 (11), 1194-1209CODEN: CDAIB2; ISSN:2159-8274. (American Association for Cancer Research)MTOR serves as a central regulator of cell growth and metab. by forming two distinct complexes, mTORC1 and mTORC2. Although mechanisms of mTORC1 activation by growth factors and amino acids have been extensively studied, the upstream regulatory mechanisms leading to mTORC2 activation remain largely elusive. Here, we report that the pleckstrin homol. (PH) domain of SIN1, an essential and unique component of mTORC2, interacts with the mTOR kinase domain to suppress mTOR activity. More importantly, PtdIns(3,4,5)P3, but not other PtdInsPn species, interacts with SIN1-PH to release its inhibition on the mTOR kinase domain, thereby triggering mTORC2 activation. Mutating crit. SIN1 residues that mediate PtdIns(3,4,5)P3 interaction inactivates mTORC2, whereas mTORC2 activity is pathol. increased by patient-derived mutations in the SIN1-PH domain, promoting cell growth and tumor formation. Together, our study unravels a PI3K-dependent mechanism for mTORC2 activation, allowing mTORC2 to activate AKT in a manner that is regulated temporally and spatially by PtdIns(3,4,5)P3. Significance: The SIN1-PH domain interacts with the mTOR kinase domain to suppress mTOR activity, and PtdIns(3,4,5)P3 binds the SIN1-PH domain to release its inhibition on the mTOR kinase domain, leading to mTORC2 activation. Cancer patient-derived SIN1-PH domain mutations gain oncogenicity by loss of suppressing mTOR activity as a means to facilitate tumorigenesis. Cancer Discov; 5(11); 1194-209. ©2015 AACR. See related commentary by Yuan and Guan, p. 1127. This article is highlighted in the In This Issue feature, p.1111.
- 26Nowak, P.; Cole, D. C.; Brooijmans, N.; Bursavich, M. G.; Curran, K. J.; Ellingboe, J. W.; Gibbons, J. J.; Hollander, I.; Hu, Y.; Kaplan, J.; Malwitz, D. J.; Toral-Barza, L.; Verheijen, J. C.; Zask, A.; Zhang, W. G.; Yu, K. Discovery of potent and selective inhibitors of the mammalian target of rapamycin (mTOR) kinase. J. Med. Chem. 2009, 52 (22), 7081– 7089, DOI: 10.1021/jm9012642Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1Oku7zL&md5=5a60157598d9eb85e4fff6dc07f01cc2Discovery of Potent and Selective Inhibitors of the Mammalian Target of Rapamycin (mTOR) KinaseNowak, Pawel; Cole, Derek C.; Brooijmans, Natasja; Curran, Kevin J.; Ellingboe, John W.; Gibbons, James J.; Hollander, Irwin; Hu, Yong Bo; Kaplan, Joshua; Malwitz, David J.; Toral-Barza, Lourdes; Verheijen, Jeroen C.; Zask, Arie; Zhang, Wei-Guo; Yu, KerJournal of Medicinal Chemistry (2009), 52 (22), 7081-7089CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The mammalian target of rapamycin (mTOR) is a central regulator of cell growth, metab., and angiogenesis and an emerging target in cancer research. High throughput screening (HTS) of our compd. collection led to the identification of 3-(4-morpholin-4-yl-1-piperidin-4-yl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenol (5a), a modestly potent and nonselective inhibitor of mTOR and phosphoinositide 3-kinase (PI3K). Optimization of compd. 5a, employing an mTOR homol. model based on an X-ray crystal structure of closely related PI3Kγ led to the discovery of 6-(1H-indol-5-yl)-4-morpholin-4-yl-1-[1-(pyridin-3-ylmethyl)piperidin-4-yl]-1H-pyrazolo[3,4-d]pyrimidine (5u), a potent and selective mTOR inhibitor (mTOR IC50 = 9 nM; PI3Kα IC50 = 1962 nM). Compd. 5u selectively inhibited cellular biomarker of mTORC1 (P-S6K, P-4EBP1) and mTORC2 (P-AKT S473) over the biomarker of PI3K/PDK1 (P-AKT T308) and did not inhibit PI3K-related kinases (PIKKs) in cellular assays. These pyrazolopyrimidines represent an exciting new series of mTOR-selective inhibitors with potential for development for cancer therapy.
- 27Jin, Z.; Niu, H.; Wang, X.; Zhang, L.; Wang, Q.; Yang, A. Preclinical study of CC223 as a potential anti-ovarian cancer agent. Oncotarget 2017, 8 (35), 58469– 58479, DOI: 10.18632/oncotarget.17753Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M%252FhtlOjuw%253D%253D&md5=091fdb75cec2ddb9018ba74c124041e8Preclinical study of CC223 as a potential anti-ovarian cancer agentJin Zhenzhen; Niu Huanfu; Wang Xuenan; Wang Qin; Yang Aijun; Zhang LeiOncotarget (2017), 8 (35), 58469-58479 ISSN:.Aberrant activation of mTOR contributes to ovarian cancer progression. CC223 is a novel and potent mTOR kinase inhibitor. The current study tested its activity against human ovarian cancer cells. We showed that CC223, at nM concentrations, inhibited survival and proliferation of established/primary human ovarian cancer cells. Further, significant apoptosis activation was observed in CC223-treated ovarian cancer cells. CC223 disrupted assembly of mTOR complex 1 (mTORC1) and mTORC2 in SKOV3 cells. Meanwhile, activation of mTORC1 and mTORC2 was almost completely blocked by CC223. Intriguingly, restoring mTOR activation by introduction of a constitutively-active Akt1 only partially inhibited CC223-induced cytotoxicity in SKOV3 cells. Further studies showed that CC223 inhibited sphingosine kinase 1 (SphK1) activity and induced reactive oxygen species (ROS) production in SKOV3 cells. At last, oral administration of CC223 potently inhibited SKOV3 xenografted tumor growth in nude mice. The results of this study imply that CC223 could be further studied as a potential anti-ovarian cancer agent.
- 28Slotkin, E. K.; Patwardhan, P. P.; Vasudeva, S. D.; de Stanchina, E.; Tap, W. D.; Schwartz, G. K. MLN0128, an ATP-competitive mTOR kinase inhibitor with potent in vitro and in vivo antitumor activity, as potential therapy for bone and soft-tissue sarcoma. Mol. Cancer Ther. 2015, 14 (2), 395– 406, DOI: 10.1158/1535-7163.MCT-14-0711Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXitlOjsbo%253D&md5=7c1d867aae6e5ca99c63b554295a539aMLN0128, an ATP-Competitive mTOR Kinase Inhibitor with Potent In Vitro and In Vivo Antitumor Activity, as Potential Therapy for Bone and Soft-Tissue SarcomaSlotkin, Emily K.; Patwardhan, Parag P.; Vasudeva, Shyamprasad D.; de Stanchina, Elisa; Tap, William D.; Schwartz, Gary K.Molecular Cancer Therapeutics (2015), 14 (2), 395-406CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that exists in two complexes (mTORC1 and mTORC2) and integrates extracellular and intracellular signals to act as a master regulator of cell growth, survival, and metab. The PI3K/AKT/mTOR prosurvival pathway is often dysregulated in multiple sarcoma subtypes. First-generation allosteric inhibitors of mTORC1 (rapalogues) have been extensively tested with great preclin. promise, but have had limited clin. utility. Here, we report that MLN0128, a second-generation, ATP-competitive, pan-mTOR kinase inhibitor, acts on both mTORC1 and mTORC2 and has potent in vitro and in vivo antitumor activity in multiple sarcoma subtypes. In vitro, MLN0128 inhibits mTORC1/2 targets in a concn.-dependent fashion and shows striking antiproliferative effect in rhabdomyosarcoma (RMS), Ewing sarcoma, malignant peripheral nerve sheath tumor, synovial sarcoma, osteosarcoma, and liposarcoma. Unlike rapamycin, MLN0128 inhibits phosphorylation of 4EBP1 and NDRG1 as well as prevents the reactivation of pAKT that occurs via neg. feedback release with mTORC1 inhibition alone. In xenograft models, MLN0128 treatment results in suppression of tumor growth with two dosing schedules (1 mg/kg daily and 3 mg/kg b.i.d. t.i.w.). At the 3 mg/kg dosing schedule, MLN0128 treatment results in significantly better tumor growth suppression than rapamycin in RMS and Ewing sarcoma models. In addn., MLN0128 induces apoptosis in models of RMS both in vitro and in vivo. Results from our study strongly suggest that MLN0128 treatment should be explored further as potential therapy for sarcoma. Mol Cancer Ther; 14(2); 395-406. ©2014 AACR.
- 29Pike, K. G.; Malagu, K.; Hummersone, M. G.; Menear, K. A.; Duggan, H. M.; Gomez, S.; Martin, N. M.; Ruston, L.; Pass, S. L.; Pass, M. Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014. Bioorg. Med. Chem. Lett. 2013, 23 (5), 1212– 1216, DOI: 10.1016/j.bmcl.2013.01.019Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFajs7c%253D&md5=325cb45f84bb4357005315ab3b27c0aaOptimization of potent and selective dual mTORC1 and mTORC2 inhibitors: The discovery of AZD8055 and AZD2014Pike, Kurt G.; Malagu, Karine; Hummersone, Marc G.; Menear, Keith A.; Duggan, Heather M. E.; Gomez, Sylvie; Martin, Niall M. B.; Ruston, Linette; Pass, Sarah L.; Pass, MartinBioorganic & Medicinal Chemistry Letters (2013), 23 (5), 1212-1216CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)The optimization of a potent and highly selective series of dual mTORC1 and mTORC2 inhibitors is described. An initial focus on improving cellular potency while maintaining or improving other key parameters, such as aq. soly. and margins over hERG IC50, led to the discovery of the clin. candidate AZD8055. Further optimization, particularly aimed at reducing the rate of metab. in human hepatocyte incubations, resulted in the discovery of the clin. candidate AZD2014.
- 30Lee, J. S.; Vo, T. T.; Fruman, D. A. Targeting mTOR for the treatment of B cell malignancies. Br. J. Clin. Pharmacol. 2016, 82 (5), 1213– 1228, DOI: 10.1111/bcp.12888Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xhs1Gqsr7L&md5=0ae355bfde2b1b275eb2bdfd55b4cbabTargeting mTOR for the treatment of B cell malignanciesLee, Jong-Hoon Scott; Vo, Thanh-Trang; Fruman, David A.British Journal of Clinical Pharmacology (2016), 82 (5), 1213-1228CODEN: BCPHBM; ISSN:1365-2125. (Wiley-Blackwell)Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that functions as a key regulator of cell growth, division and survival. Many haematol. malignancies exhibit elevated or aberrant mTOR activation, supporting the launch of numerous clin. trials aimed at evaluating the potential of single agent mTOR-targeted therapies. While promising early clin. data using allosteric mTOR inhibitors (rapamycin and its derivs., rapalogs) have suggested activity in a subset of haematol. malignancies, these agents have shown limited efficacy in most contexts. Whether the efficacy of these partial mTOR inhibitors might be enhanced by more complete target inhibition is being actively addressed with second generation ATP-competitive mTOR kinase inhibitors (TOR-KIs), which have only recently entered clin. trials. However, emerging preclin. data suggest that despite their biochem. advantage over rapalogs, TOR-KIs may retain a primarily cytostatic response. Rather, combinations of mTOR inhibition with other targeted therapies have demonstrated promising efficacy in several preclin. models. This review investigates the current status of rapalogs and TOR-KIs in B cell malignancies, with an emphasis on emerging preclin. evidence of synergistic combinations involving mTOR inhibition.
- 31Rageot, D.; Bohnacker, T.; Melone, A.; Langlois, J. B.; Borsari, C.; Hillmann, P.; Sele, A. M.; Beaufils, F.; Zvelebil, M.; Hebeisen, P.; Loscher, W.; Burke, J.; Fabbro, D.; Wymann, M. P. Discovery and preclinical characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoro methyl)pyridin-2-amine (PQR620), a highly potent and selective mTORC1/2 inhibitor for cancer and neurological disorders. J. Med. Chem. 2018, 61 (22), 10084– 10105, DOI: 10.1021/acs.jmedchem.8b01262Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvF2mtLjO&md5=e6ebf8147d6ba041285eb6538ade46abDiscovery and Preclinical Characterization of 5-[4,6-Bis({3-oxa-8-azabicyclo[3.2.1]octan-8-yl})-1,3,5-triazin-2-yl]-4-(difluoromethyl)pyridin-2-amine (PQR620), a Highly Potent and Selective mTORC1/2 Inhibitor for Cancer and Neurological DisordersRageot, Denise; Bohnacker, Thomas; Melone, Anna; Langlois, Jean-Baptiste; Borsari, Chiara; Hillmann, Petra; Sele, Alexander M.; Beaufils, Florent; Zvelebil, Marketa; Hebeisen, Paul; Loscher, Wolfgang; Burke, John; Fabbro, Doriano; Wymann, Matthias P.Journal of Medicinal Chemistry (2018), 61 (22), 10084-10105CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Mechanistic target of rapamycin (mTOR) promotes cell proliferation, growth, and survival and is overactivated in many tumors and central nervous system disorders. I is a novel, potent, selective, and brain penetrable inhibitor of mTORC1/2 kinase. I showed excellent selectivity for mTOR over PI3K and protein kinases and efficiently prevented cancer cell growth in a 66 cancer cell line panel. In C57BL/6J and Sprague-Dawley mice, max. concn. (Cmax) in plasma and brain was reached after 30 min, with a half-life (t1/2) > 5 h. In an ovarian carcinoma mouse xenograft model (OVCAR-3), daily dosing of I inhibited tumor growth significantly. Moreover, I attenuated epileptic seizures in a tuberous sclerosis complex (TSC) mouse model. In conclusion, I inhibits mTOR kinase potently and selectively, shows antitumor effects in vitro and in vivo, and promises advantages in CNS indications due to its brain/plasma distribution ratio.
- 32Bohnacker, T.; Prota, A. E.; Beaufils, F.; Burke, J. E.; Melone, A.; Inglis, A. J.; Rageot, D.; Sele, A. M.; Cmiljanovic, V.; Cmiljanovic, N.; Bargsten, K.; Aher, A.; Akhmanova, A.; Diaz, J. F.; Fabbro, D.; Zvelebil, M.; Williams, R. L.; Steinmetz, M. O.; Wymann, M. P. Deconvolution of Buparlisib’s mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat. Commun. 2017, 8, 14683, DOI: 10.1038/ncomms14683Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1czksV2ruw%253D%253D&md5=97d27a7d83bf6ab6094a587565f82bf6Deconvolution of Buparlisib's mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic interventionBohnacker Thomas; Beaufils Florent; Melone Anna; Rageot Denise; Sele Alexander M; Cmiljanovic Vladimir; Cmiljanovic Natasa; Wymann Matthias P; Prota Andrea E; Bargsten Katja; Steinmetz Michel O; Burke John E; Inglis Alison J; Williams Roger L; Aher Amol; Akhmanova Anna; Diaz J Fernando; Fabbro Doriano; Zvelebil MarketaNature communications (2017), 8 (), 14683 ISSN:.BKM120 (Buparlisib) is one of the most advanced phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of cancer, but it interferes as an off-target effect with microtubule polymerization. Here, we developed two chemical derivatives that differ from BKM120 by only one atom. We show that these minute changes separate the dual activity of BKM120 into discrete PI3K and tubulin inhibitors. Analysis of the compounds cellular growth arrest phenotypes and microtubule dynamics suggest that the antiproliferative activity of BKM120 is mainly due to microtubule-dependent cytotoxicity rather than through inhibition of PI3K. Crystal structures of BKM120 and derivatives in complex with tubulin and PI3K provide insights into the selective mode of action of this class of drugs. Our results raise concerns over BKM120's generally accepted mode of action, and provide a unique mechanistic basis for next-generation PI3K inhibitors with improved safety profiles and flexibility for use in combination therapies.
- 33Beaufils, F.; Cmiljanovic, N.; Cmiljanovic, V.; Bohnacker, T.; Melone, A.; Marone, R.; Jackson, E.; Zhang, X.; Sele, A.; Borsari, C.; Mestan, J.; Hebeisen, P.; Hillmann, P.; Giese, B.; Zvelebil, M.; Fabbro, D.; Williams, R. L.; Rageot, D.; Wymann, M. P. 5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a potent, brain-penetrant, orally bioavailable, pan-class I PI3K/mTOR inhibitor as clinical candidate in oncology. J. Med. Chem. 2017, 60 (17), 7524– 7538, DOI: 10.1021/acs.jmedchem.7b00930Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtlKhurrP&md5=54d4db75f6a8bda3f12eaf1d196352fd5-(4,6-Dimorpholino-1,3,5-triazin-2-yl)-4-(trifluoromethyl)pyridin-2-amine (PQR309), a Potent, Brain-Penetrant, Orally Bioavailable, Pan-Class I PI3K/mTOR Inhibitor as Clinical Candidate in OncologyBeaufils, Florent; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Bohnacker, Thomas; Melone, Anna; Marone, Romina; Jackson, Eileen; Zhang, Xuxiao; Sele, Alexander; Borsari, Chiara; Mestan, Jurgen; Hebeisen, Paul; Hillmann, Petra; Giese, Bernd; Zvelebil, Marketa; Fabbro, Doriano; Williams, Roger L.; Rageot, Denise; Wymann, Matthias P.Journal of Medicinal Chemistry (2017), 60 (17), 7524-7538CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Phosphoinositide 3-kinase (PI3K) is deregulated in a wide variety of human tumors and triggers activation of protein kinase B (PKB/Akt) and mammalian target of rapamycin (mTOR). Here we describe the preclin. characterization of compd. 1 (PQR309, bimiralisib), a potent 4,6-dimorpholino-1,3,5-triazine-based pan-class I PI3K inhibitor, which targets mTOR kinase in a balanced fashion at higher concns. No off-target interactions were detected for 1 in a wide panel of protein kinase, enzyme, and receptor ligand assays. Moreover, 1 did not bind tubulin, which was obsd. for the structurally related 4 (BKM120, buparlisib). Compd. 1 is orally available, crosses the blood-brain barrier, and displayed favorable pharmacokinetic parameters in mice, rats, and dogs. Compd. 1 demonstrated efficiency in inhibiting proliferation in tumor cell lines and a rat xenograft model. This, together with the compd.'s safety profile, identifies 1 as a clin. candidate with a broad application range in oncol., including treatment of brain tumors or CNS metastasis. Compd. 1 is currently in phase II clin. trials for advanced solid tumors and refractory lymphoma.
- 34Tarantelli, C.; Gaudio, E.; Arribas, A. J.; Kwee, I.; Hillmann, P.; Rinaldi, A.; Cascione, L.; Spriano, F.; Bernasconi, E.; Guidetti, F.; Carrassa, L.; Pittau, R. B.; Beaufils, F.; Ritschard, R.; Rageot, D.; Sele, A.; Dossena, B.; Rossi, F. M.; Zucchetto, A.; Taborelli, M.; Gattei, V.; Rossi, D.; Stathis, A.; Stussi, G.; Broggini, M.; Wymann, M. P.; Wicki, A.; Zucca, E.; Cmiljanovic, V.; Fabbro, D.; Bertoni, F. PQR309 is a novel dual PI3K/mTOR inhibitor with preclinical antitumor activity in lymphomas as a single agent and in combination therapy. Clin. Cancer Res. 2018, 24 (1), 120– 129, DOI: 10.1158/1078-0432.CCR-17-1041Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVWktw%253D%253D&md5=730305c33b7f669b0eedd0614edd886cPQR309 Is a Novel Dual PI3K/mTOR Inhibitor with Preclinical Antitumor Activity in Lymphomas as a Single Agent and in Combination TherapyTarantelli, Chiara; Gaudio, Eugenio; Arribas, Alberto J.; Kwee, Ivo; Hillmann, Petra; Rinaldi, Andrea; Cascione, Luciano; Spriano, Filippo; Bernasconi, Elena; Guidetti, Francesca; Carrassa, Laura; Pittau, Roberta Bordone; Beaufils, Florent; Ritschard, Reto; Rageot, Denise; Sele, Alexander; Dossena, Barbara; Rossi, Francesca Maria; Zucchetto, Antonella; Taborelli, Monica; Gattei, Valter; Rossi, Davide; Stathis, Anastasios; Stussi, Georg; Broggini, Massimo; Wymann, Matthias P.; Wicki, Andreas; Zucca, Emanuele; Cmiljanovic, Vladimir; Fabbro, Doriano; Bertoni, FrancescoClinical Cancer Research (2018), 24 (1), 120-129CODEN: CCREF4; ISSN:1078-0432. (American Association for Cancer Research)Purpose: Activation of the PI3K/mTOR signaling pathway is recurrent in different lymphoma types, and pharmacol. inhibition of the PI3K/mTOR pathway has shown activity in lymphoma patients. Here, we extensively characterized the in vitro and in vivo activity and the mechanism of action of PQR309 (bimiralisib), a novel oral selective dual PI3K/mTOR inhibitor under clin. evaluation, in preclin. lymphoma models. Exptl. Design: This study included preclin. in vitro activity screening on a large panel of cell lines, both as single agent and in combination, validation expts. on in vivo models and primary cells, proteomics and gene-expression profiling, and comparison with other signaling inhibitors. Results: PQR309 had in vitro antilymphoma activity as single agent and in combination with venetoclax, panobinostat, ibrutinib, lenalidomide, ARV-825, marizomib, and rituximab. Sensitivity to PQR309 was assocd. with specific baseline gene-expression features, such as high expression of transcripts coding for the BCR pathway. Combining proteomics and RNA profiling, we identified the different contribution of PQR309-induced protein phosphorylation and gene expression changes to the drug mechanism of action. Gene-expression signatures induced by PQR309 and by other signaling inhibitors largely overlapped. PQR309 showed activity in cells with primary or secondary resistance to idelalisib. Conclusions: On the basis of these results, PQR309 appeared as a novel and promising compd. that is worth developing in the lymphoma setting. Clin Cancer Res; 24(1); 120-9. ©2017 AACR.
- 35Wicki, A.; Brown, N.; Xyrafas, A.; Bize, V.; Hawle, H.; Berardi, S.; Cmiljanovic, N.; Cmiljanovic, V.; Stumm, M.; Dimitrijevic, S.; Herrmann, R.; Pretre, V.; Ritschard, R.; Tzankov, A.; Hess, V.; Childs, A.; Hierro, C.; Rodon, J.; Hess, D.; Joerger, M.; von Moos, R.; Sessa, C.; Kristeleit, R. First-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13). Eur. J. Cancer 2018, 96, 6– 16, DOI: 10.1016/j.ejca.2018.03.012Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Snu70%253D&md5=acbdfdb59498f5543ba7f02ed9709d0eFirst-in human, phase 1, dose-escalation pharmacokinetic and pharmacodynamic study of the oral dual PI3K and mTORC1/2 inhibitor PQR309 in patients with advanced solid tumors (SAKK 67/13)Wicki, Andreas; Brown, Nicholas; Xyrafas, Alexandros; Bize, Vincent; Hawle, Hanne; Berardi, Simona; Cmiljanovic, Natasa; Cmiljanovic, Vladimir; Stumm, Michael; Dimitrijevic, Sasa; Herrmann, Richard; Pretre, Vincent; Ritschard, Reto; Tzankov, Alexandar; Hess, Viviane; Childs, Alexa; Hierro, Cinta; Rodon, Jordi; Hess, Dagmar; Joerger, Markus; von Moos, Roger; Sessa, Cristiana; Kristeleit, RebeccaEuropean Journal of Cancer (2018), 96 (), 6-16CODEN: EJCAEL; ISSN:0959-8049. (Elsevier Ltd.)PQR309 is an orally bioavailable, balanced pan-phosphatidylinositol-3-kinase (PI3K), mammalian target of rapamycin (mTOR) C1 and mTORC2 inhibitor. This is an accelerated titrn., 3 D 3 dose-escalation, open-label phase Itrial of continuous once-daily (OD) PQR309 administration to evaluate the safety, pharmacokinetics (PK) and pharmacodynamics in patients with advanced solid tumors. Primary objectives were to det. the max. tolerated dose (MTD) and recommended phase 2 dose (RP2D).Twenty-eight patients were included in six dosing cohorts and treated at a daily PQR309 dose ranging from 10 to 150 mg. Common adverse events (AEs; ≥30% patients) included fatigue, hyperglycemia, nausea, diarrhea, constipation, rash, anorexia and vomiting. Grade (G) 3 or 4 drug-related AEs were seen in 13 (46%) and three (11%) patients, resp. Dose-limiting toxicity (DLT) was obsd. in two patients at 100 mg OD (>14-d interruption in PQR309 due to G3 rash, G2 hyperbilirubinemia, G4 suicide attempt; dose redn. due to G3 fatigue, G2 diarrhoea, G4 transaminitis) and one patient at 80 mg (G3 hyperglycemia >7 d). PK shows fast absorption (Tmax 1-2 h) and dose proportionality for Cmax and area under the curve. A partial response in a patient with metastatic thymus cancer, 24% disease vol. redn. in a patient with sinonasal cancer and stable disease for more than 16 wk in a patient with clear cell Bartholin's gland cancer were obsd.The MTD and RP2D of PQR309 is 80 mg of orally OD. PK is dose-proportional. PD shows PI3K pathway phosphoprotein downregulation in paired tumor biopsies. Clin. activity was obsd. in patients with and without PI3K pathway dysregulation.
- 36Fang, Z.; Song, Y.; Zhan, P.; Zhang, Q.; Liu, X. Conformational restriction: an effective tactic in “follow-on”-based drug discovery. Future Med. Chem. 2014, 6 (8), 885– 901, DOI: 10.4155/fmc.14.50Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVemsrzE&md5=dcf5fa6cead84c77b3603c7e0800f49bConformational restriction: an effective tactic in 'follow-on'-based drug discoveryFang, Zengjun; Song, Yu'ning; Zhan, Peng; Zhang, Qingzhu; Liu, XinyongFuture Medicinal Chemistry (2014), 6 (8), 885-901CODEN: FMCUA7; ISSN:1756-8919. (Future Science Ltd.)A review. The conformational restriction (rigidification) of a flexible ligand has often been a commonly used strategy in drug design, as it can minimize the entropic loss assocd. with the ligand adopting a preferred conformation for binding, which leads to enhanced potency for a given physiol. target, improved selectivity for isoforms and reduced the possibility of drug metab. Therefore, the application of conformational restriction strategy is a core aspect of drug discovery and development that is widely practiced by medicinal chemists either deliberately or subliminally. The present review will highlight current representative examples and a brief overview on the rational design of conformationally restricted agents as well as discuss its advantages over the flexible counterparts.
- 37Cmiljanovic, V.; Hebeisen, P.; Jackson, E.; Beaufils, F.; Bohnacker, T.; Wymann, M. P. Conformationally Restricted PI3K and mTOR Inhibitors. Patent WO2015049369, 2015.Google ScholarThere is no corresponding record for this reference.
- 38Leroux, F. R.; Manteau, B.; Vors, J. P.; Pazenok, S. Trifluoromethyl ethers--synthesis and properties of an unusual substituent. Beilstein J. Org. Chem. 2008, 4 (13), DOI: 10.3762/bjoc.4.13 .Google ScholarThere is no corresponding record for this reference.
- 39Burger, M. T.; Pecchi, S.; Wagman, A.; Ni, Z. J.; Knapp, M.; Hendrickson, T.; Atallah, G.; Pfister, K.; Zhang, Y.; Bartulis, S.; Frazier, K.; Ng, S.; Smith, A.; Verhagen, J.; Haznedar, J.; Huh, K.; Iwanowicz, E.; Xin, X.; Menezes, D.; Merritt, H.; Lee, I.; Wiesmann, M.; Kaufman, S.; Crawford, K.; Chin, M.; Bussiere, D.; Shoemaker, K.; Zaror, I.; Maira, S. M.; Voliva, C. F. Identification of NVP-BKM120 as a potent, selective, orally bioavailable class I PI3 Kinase inhibitor for treating cancer. ACS Med. Chem. Lett. 2011, 2 (10), 774– 779, DOI: 10.1021/ml200156tGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtVOktrrI&md5=5d00d73dda1fc0d8f0f07d12900282edIdentification of NVP-BKM120 as a Potent, Selective, Orally Bioavailable Class I PI3 Kinase Inhibitor for Treating CancerBurger, Matthew T.; Pecchi, Sabina; Wagman, Allan; Ni, Zhi-Jie; Knapp, Mark; Hendrickson, Thomas; Atallah, Gordana; Pfister, Keith; Zhang, Yanchen; Bartulis, Sarah; Frazier, Kelly; Ng, Simon; Smith, Aaron; Verhagen, Joelle; Haznedar, Joshua; Huh, Kay; Iwanowicz, Ed; Xin, Xiaohua; Menezes, Daniel; Merritt, Hanne; Lee, Isabelle; Wiesmann, Marion; Kaufman, Susan; Crawford, Kenneth; Chin, Michael; Bussiere, Dirksen; Shoemaker, Kevin; Zaror, Isabel; Maira, Sauveur-Michel; Voliva, Charles F.ACS Medicinal Chemistry Letters (2011), 2 (10), 774-779CODEN: AMCLCT; ISSN:1948-5875. (American Chemical Society)Phosphoinositide-3-kinases (PI3Ks) are important oncol. targets due to the deregulation of this signaling pathway in a wide variety of human cancers. Herein we describe the structure guided optimization of a series of 2-morpholino, 4-substituted, 6-heterocyclic pyrimidines where the pharmacokinetic properties were improved by modulating the electronics of the 6-position heterocycle, and the overall druglike properties were fine-tuned further by modification of the 4-position substituent. The resulting 2,4-bismorpholino 6-heterocyclic pyrimidines are potent class I PI3K inhibitors showing mechanism modulation in PI3K dependent cell lines and in vivo efficacy in tumor xenograft models with PI3K pathway deregulation (A2780 ovarian and U87MG glioma). These efforts culminated in the discovery of 15 (NVP-BKM120), currently in Phase II clin. trials for the treatment of cancer.
- 40Rousseau, J. F.; Chekroun, I.; Ferey, V.; Labrosse, J. R. Concise preparation of a stable cyclic sulfamidate intermediate in the synthesis of a enantiopure chiral active diamine derivative. Org. Process Res. Dev. 2015, 19 (4), 506– 513, DOI: 10.1021/op500264vGoogle Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXksVOrurk%253D&md5=72c5ea832b893e2bccf01a80c9ea1734Concise Preparation of a Stable Cyclic Sulfamidate Intermediate in the Synthesis of a Enantiopure Chiral Active Diamine DerivativeRousseau, Jean-Francois; Chekroun, Isaac; Ferey, Vincent; Labrosse, Jean RobertOrganic Process Research & Development (2015), 19 (4), 506-513CODEN: OPRDFK; ISSN:1083-6160. (American Chemical Society)A potentially scalable route to the nonracemic antipsychotic candidate SSR 504374 I was developed using the stereoselective substitution reaction of nonracemic sulfamidate II with 2-chloro-3-trifluoromethylbenzamide as the key step. II was prepd. in seven steps from 2-benzoylpyridine by chemo- and diastereoselective hydrogenation, sepn. of the desired racemic erythro diastereomer, resoln. with di-p-toluoyl-(+)-tartaric acid, formation of a sulfamidite, and oxidn. at sulfur with RuCl3 and sodium hypochlorite; a workup for the sulfamidite oxidn. using isopropanol was developed to avoid darkening of the intermediate due to oxidn. by RuO2 and RuO4 left in the sulfamidate product.
- 41Brown, G. R.; Foubister, A. J.; Wright, B. Chiral synthesis of 3-substituted morpholines via serine enantiomers and reductions of 5-oxomorpholine-3-carboxylates. J. Chem. Soc., Perkin Trans. 1 1985, 1, 2577– 2580, DOI: 10.1039/p19850002577Google ScholarThere is no corresponding record for this reference.
- 42Hebeisen, P.; Alker, A.; Buerkler, M. Iterative one pot reactions of a chiral sulfamidate with 2,4,6-trichloropyridine: regiocontrolled synthesis of linear and angular chiral dipyrrolidino pyridines. Heterocycles 2012, 85 (1), 65– 72, DOI: 10.3987/COM-11-12360Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XpslWnug%253D%253D&md5=07450591f843628878b047a25205e5bfIterative one pot reactions of a chiral sulfamidate with 2,4,6-trichloropyridine: Regiocontrolled synthesis of linear and angular chiral dipyrrolidino pyridinesHebeisen, Paul; Alker, Andre; Buerkler, MarkusHeterocycles (2012), 85 (1), 65-72CODEN: HTCYAM; ISSN:0385-5414. (Japan Institute of Heterocyclic Chemistry)The product of the ring opening of a chiral sulfamidate with the 3-lithiopyridine species obtained by deprotonation of 2,4,6-trichloropyridine with BuLi was deprotonated again in situ with BuLi and reacted with a 2nd equiv. of the sulfamidate furnishing a bis(β-aminoethyl)pyridine deriv., which could be cyclized regioselectively to linear or angular chiral dipyrrolidinopyridines.
- 43Zask, A.; Kaplan, J.; Verheijen, J. C.; Richard, D. J.; Curran, K.; Brooijmans, N.; Bennett, E. M.; Toral-Barza, L.; Hollander, I.; Ayral-Kaloustian, S.; Yu, K. Morpholine derivatives greatly enhance the selectivity of mammalian target of rapamycin (mTOR) inhibitors. J. Med. Chem. 2009, 52 (24), 7942– 7945, DOI: 10.1021/jm901415xGoogle Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVWrtb3J&md5=5ef1eee5aa79531970049d3d26b46eb2Morpholine Derivatives Greatly Enhance the Selectivity of Mammalian Target of Rapamycin (mTOR) InhibitorsZask, Arie; Kaplan, Joshua; Verheijen, Jeroen C.; Richard, David J.; Curran, Kevin; Brooijmans, Natasja; Bennett, Eric M.; Toral-Barza, Lourdes; Hollander, Irwin; Ayral-Kaloustian, Semiramis; Yu, KerJournal of Medicinal Chemistry (2009), 52 (24), 7942-7945CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Dramatic improvements in mTOR-targeting selectivity were achieved by replacing morpholine in pyrazolopyrimidine inhibitors with bridged morpholines. Analogs I [R1 = (R)- or (S)-3-methyl-4-morpholinyl, 2-methyl-4-morpholinyl, 8-oxa-3-azabicyclo[3.2.1]octan-3-yl, etc.; R2 = Me, Et, cyclopropyl, FCH2CH2, 3-pyridyl, 4-pyridyl; R3 = F3CCH2, 1-methoxycarbonyl-4-piperidinyl, 1-ethoxycarbonyl-4-piperidinyl, 3-pyridylmethyl] with subnanomolar mTOR IC50 values and up to 26000-fold selectivity vs. PI3Kα were prepd. Chiral morpholines gave inhibitors whose enantiomers had different selectivity and potency profiles. Mol. modeling suggests that a single amino acid difference between PI3K and mTOR (Phe961Leu) accounts for the profound selectivity seen by creating a deeper pocket in mTOR that can accommodate bridged morpholines.
- 44Thomas, V. H.; Bhattachar, S.; Hitchingham, L.; Zocharski, P.; Naath, M.; Surendran, N.; Stoner, C. L.; El-Kattan, A. The road map to oral bioavailability: an industrial perspective. Expert Opin. Drug Metab. Toxicol. 2006, 2 (4), 591– 608, DOI: 10.1517/17425255.2.4.591Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnt1ahsLs%253D&md5=79cf4cea1b89914bd3a8826b5d7360f9The road map to oral bioavailability: an industrial perspectiveThomas, V. Hayden; Bhattachar, Shobha; Hitchingham, Linda; Zocharski, Philip; Naath, Maryanne; Surendran, Narayanan; Stoner, Chad L.; El-Kattan, AymanExpert Opinion on Drug Metabolism & Toxicology (2006), 2 (4), 591-608CODEN: EODMAP; ISSN:1742-5255. (Informa Healthcare)A review. Optimization of oral bioavailability is a continuing challenge for the pharmaceutical and biotechnol. industries. The no. of potential drug candidates requiring in vivo evaluation has significantly increased with the advent of combinatorial chem. In addn., drug discovery programs are increasingly forced into more lipophilic and lower soly. chem. space. To aid in the use of in vitro and in silico tools as well as reduce the no. of in vivo studies required, a team-based discussion tool is proposed that provides a road map' to guide the selection of profiling assays that should be considered when optimizing oral bioavailability. This road map divides the factors that contribute to poor oral bioavailability into two interrelated categories: absorption and metab. This road map provides an interface for cross discipline discussions and a systematic approach to the experimentation that drives the drug discovery process towards a common goal - acceptable oral bioavailability using minimal resources in an acceptable time frame.
- 45Mortensen, D. S.; Fultz, K. E.; Xu, S.; Xu, W.; Packard, G.; Khambatta, G.; Gamez, J. C.; Leisten, J.; Zhao, J.; Apuy, J.; Ghoreishi, K.; Hickman, M.; Narla, R. K.; Bissonette, R.; Richardson, S.; Peng, S. X.; Perrin-Ninkovic, S.; Tran, T.; Shi, T.; Yang, W. Q.; Tong, Z.; Cathers, B. E.; Moghaddam, M. F.; Canan, S. S.; Worland, P.; Sankar, S.; Raymon, H. K. CC-223, a potent and selective inhibitor of mTOR kinase: in vitro and in vivo characterization. Mol. Cancer Ther. 2015, 14 (6), 1295– 1305, DOI: 10.1158/1535-7163.MCT-14-1052Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXpsFOgurk%253D&md5=f33b21dc89dedaf14fab54a819afb917CC-223, a Potent and Selective Inhibitor of mTOR Kinase: In Vitro and In Vivo CharacterizationMortensen, Deborah S.; Fultz, Kimberly E.; Xu, Shuichan; Xu, Weiming; Packard, Garrick; Khambatta, Godrej; Gamez, James C.; Leisten, Jim; Zhao, Jingjing; Apuy, Julius; Ghoreishi, Kamran; Hickman, Matt; Narla, Rama Krishna; Bissonette, Rene; Richardson, Samantha; Peng, Sophie X.; Perrin-Ninkovic, Sophie; Tran, Tam; Shi, Tao; Yang, Wen Qing; Tong, Zeen; Cathers, Brian E.; Moghaddam, Mehran F.; Canan, Stacie S.; Worland, Peter; Sankar, Sabita; Raymon, Heather K.Molecular Cancer Therapeutics (2015), 14 (6), 1295-1305CODEN: MCTOCF; ISSN:1535-7163. (American Association for Cancer Research)MTOR is a serine/threonine kinase that regulates cell growth, metab., proliferation, and survival. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are crit. mediators of the PI3K-AKT pathway, which is frequently mutated in many cancers, leading to hyperactivation of mTOR signaling. Although rapamycin analogs, allosteric inhibitors that target only the mTORC1 complex, have shown some clin. activity, it is hypothesized that mTOR kinase inhibitors, blocking both mTORC1 and mTORC2 signaling, will have expanded therapeutic potential. Here, we describe the preclin. characterization of CC-223. CC-223 is a potent, selective, and orally bioavailable inhibitor of mTOR kinase, demonstrating inhibition of mTORC1 (pS6RP and p4EBP1) and mTORC2 [pAKT(S473)] in cellular systems. Growth inhibitory activity was demonstrated in hematol. and solid tumor cell lines. mTOR kinase inhibition in cells, by CC-223, resulted in more complete inhibition of the mTOR pathway biomarkers and improved antiproliferative activity as compared with rapamycin. Growth inhibitory activity and apoptosis was demonstrated in a panel of hematol. cancer cell lines. Correlative anal. revealed that IRF4 expression level assocs. with resistance, whereas mTOR pathway activation seems to assoc. with sensitivity. Treatment with CC-223 afforded in vivo tumor biomarker inhibition in tumor-bearing mice, after a single oral dose. CC-223 exhibited dose-dependent tumor growth inhibition in multiple solid tumor xenografts. Significant inhibition of mTOR pathway markers pS6RP and pAKT in CC-223-treated tumors suggests that the obsd. antitumor activity of CC-223 was mediated through inhibition of both mTORC1 and mTORC2. CC-223 is currently in phase I clin. trials. Mol Cancer Ther; 14(6); 1295-305. ©2015 AACR.
- 46Nosik, P. S.; Ryabukhin, S. V.; Artamonov, O. S.; Grygorenko, O. O. Synthesis of trans-disubstituted pyrazolylcyclopropane building blocks. Monatsh. Chem. 2016, 147 (9), 1629– 1636, DOI: 10.1007/s00706-016-1726-6Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmsFyksbg%253D&md5=1b4cd76fed0aa27fc3134d8bbe4a301fSynthesis of trans-disubstituted pyrazolylcyclopropane building blocksNosik, Pavel S.; Ryabukhin, Sergey V.; Artamonov, Oleksiy S.; Grygorenko, Oleksandr O.Monatshefte fuer Chemie (2016), 147 (9), 1629-1636CODEN: MOCMB7; ISSN:0026-9247. (Springer-Verlag GmbH)Diastereoselective synthesis of trans-disubstituted pyrazolylcyclopropane building blocks (i.e. carboxylic acids and amines) is described starting from easily available pyrazolecarbaldehydes. The key step of the synthesis was Corey-Chaikowsky cyclopropanation of the corresponding α,β-unsatd. Weinreb amides. The title compds. were prepd. in four or six steps and 32-60 and 17-40 % overall yields, resp., on up to 50 g scale. The building blocks obtained are good starting points for the design of lead-like libraries of peptidomimetic drugs.
- 47Zhang, L.; Luo, S.; Mi, X.; Liu, S.; Qiao, Y.; Xu, H.; Cheng, J. P. Combinatorial synthesis of functionalized chiral and doubly chiral ionic liquids and their applications as asymmetric covalent/non-covalent bifunctional organocatalysts. Org. Biomol. Chem. 2008, 6 (3), 567– 576, DOI: 10.1039/B713843AGoogle Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtVWhs7o%253D&md5=4cec4a7bff2e3b65c7aac57156230d27Combinatorial synthesis of functionalized chiral and doubly chiral ionic liquids and their applications as asymmetric covalent/non-covalent bifunctional organocatalystsZhang, Long; Luo, Sanzhong; Mi, Xueling; Liu, Song; Qiao, Yupu; Xu, Hui; Cheng, Jin-PeiOrganic & Biomolecular Chemistry (2008), 6 (3), 567-576CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)A facile combinatorial strategy was developed for the construction of libraries of functionalized chiral ionic liqs. (FCILs) including doubly chiral ionic liqs. and bis-functional chiral ionic liqs. These FCIL libraries have the potential to be used as asym. catalysts or chiral ligands. As an example, novel asym. bifunctional catalysts were developed by simultaneously incorporating functional groups onto the cation and anion. The resultant bis-functionalized CILs showed significantly improved stereoselectivity over the mono-functionalized parent CILs.
- 48Fabian, M. A.; Biggs, W. H.; Treiber, D. K.; Atteridge, C. E.; Azimioara, M. D.; Benedetti, M. G.; Carter, T. A.; Ciceri, P.; Edeen, P. T.; Floyd, M.; Ford, J. M.; Galvin, M.; Gerlach, J. L.; Grotzfeld, R. M.; Herrgard, S.; Insko, D. E.; Insko, M. A.; Lai, A. G.; Lélias, J. M.; Mehta, S. A.; Milanov, Z. V.; Velasco, A. M.; Wodicka, L. M.; Patel, H. K.; Zarrinkar, P. P.; Lockhart, D. J. A small molecule-kinase interaction map for clinical kinase inhibitors. Nat. Biotechnol. 2005, 23, 329– 336, DOI: 10.1038/nbt1068Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXitF2nt7w%253D&md5=6aedd5ceb8f77cd26ee50425dcec7bdfA small molecule-kinase interaction map for clinical kinase inhibitorsFabian, Miles A.; Biggs, William H.; Treiber, Daniel K.; Atteridge, Corey E.; Azimioara, Mihai D.; Benedetti, Michael G.; Carter, Todd A.; Ciceri, Pietro; Edeen, Philip T.; Floyd, Mark; Ford, Julia M.; Galvin, Margaret; Gerlach, Jay L.; Grotzfeld, Robert M.; Herrgard, Sanna; Insko, Darren E.; Insko, Michael A.; Lai, Andiliy G.; Lelias, Jean-Michel; Mehta, Shamal A.; Milanov, Zdravko V.; Velasco, Anne Marie; Wodicka, Lisa M.; Patel, Hitesh K.; Zarrinkar, Patrick P.; Lockhart, David J.Nature Biotechnology (2005), 23 (3), 329-336CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Kinase inhibitors show great promise as a new class of therapeutics. Here the authors describe an efficient way to det. kinase inhibitor specificity by measuring binding of small mols. to the ATP site of kinases. The authors have profiled 20 kinase inhibitors, including 16 that are approved drugs or in clin. development, against a panel of 119 protein kinases. The authors find that specificity varies widely and is not strongly correlated with chem. structure or the identity of the intended target. Many novel interactions were identified, including tight binding of the p38 inhibitor BIRB-796 to an imatinib-resistant variant of the ABL kinase, and binding of imatinib to the SRC-family kinase LCK. The authors also show that mutations in the epidermal growth factor receptor (EGFR) found in gefitinib-responsive patients do not affect the binding affinity of gefitinib or erlotinib. Our results represent a systematic small mol.-protein interaction map for clin. compds. across a large no. of related proteins.
- 49Karaman, M. W.; Herrgard, S.; Treiber, D. K.; Gallant, P.; Atteridge, C. E.; Campbell, B. T.; Chan, K. W.; Ciceri, P.; Davis, M. I.; Edeen, P. T.; Faraoni, R.; Floyd, M.; Hunt, J. P.; Lockhart, D. J.; Milanov, Z. V.; Morrison, M. J.; Pallares, G.; Patel, H. K.; Pritchard, S.; Wodicka, L. M.; Zarrinkar, P. P. A quantitative analysis of kinase inhibitor selectivity. Nat. Biotechnol. 2008, 26, 127– 132, DOI: 10.1038/nbt1358Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXisFGlsQ%253D%253D&md5=346265d412853ced636ad4128ed8a76fA quantitative analysis of kinase inhibitor selectivityKaraman, Mazen W.; Herrgard, Sanna; Treiber, Daniel K.; Gallant, Paul; Atteridge, Corey E.; Campbell, Brian T.; Chan, Katrina W.; Ciceri, Pietro; Davis, Mindy I.; Edeen, Philip T.; Faraoni, Raffaella; Floyd, Mark; Hunt, Jeremy P.; Lockhart, Daniel J.; Milanov, Zdravko V.; Morrison, Michael J.; Pallares, Gabriel; Patel, Hitesh K.; Pritchard, Stephanie; Wodicka, Lisa M.; Zarrinkar, Patrick P.Nature Biotechnology (2008), 26 (1), 127-132CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Kinase inhibitors are a new class of therapeutics with a propensity to inhibit multiple targets. The biol. consequences of multikinase activity are poorly defined, and an important step toward understanding the relationship between selectivity, efficacy and safety is the exploration of how inhibitors interact with the human kinome. The authors present interaction maps for 38 kinase inhibitors across a panel of 317 kinases representing >50% of the predicted human protein kinome. The data constitute the most comprehensive study of kinase inhibitor selectivity to date and reveal a wide diversity of interaction patterns. To enable a global anal. of the results, the authors introduce the concept of a selectivity score as a general tool to quantify and differentiate the obsd. interaction patterns. The authors further investigate the impact of panel size and find that small assay panels do not provide a robust measure of selectivity.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acs.jmedchem.9b00972.
Syntheses of bromo derivative 44 (Scheme S1), bromo derivative 49 (Scheme S2), and compounds 3a–5a and 3b–5b (Scheme S3); plasma concentration of 7b after a single po dose of 5 mg/kg in rats (Table S1); brain concentration of 7b after a single po dose of 5 mg/kg in rats (Table S2); plasma concentration of 12b after a single po dose of 5 mg/kg in rats (Table S3); brain concentration of 12b after a single po dose of 5 mg/kg in rats (Table S4); stability of compound 11b (5 μM) in primary mouse, rat, dog, and human hepatocytes (Table S5); CYP1A1 and CYP1A2 metabolites identification of 11b (Table S6); proposed metabolic pathway for CYP-dependent metabolism of 11b (Figure S1); chromatogram of compound 11b incubated with CYP1A1 (60 min) (Figure S2); chromatogram of compound 11b incubated with CYP1A1 (0 min) (Figure S3); chromatogram of compound 11b incubated with CYP1A2 (60 min) (Figure S4); chromatogram of compound 11b incubated with CYP1A2 (0 min) (Figure S5); activity data and standard errors of final compounds (Table S7); activity data and standard errors of compounds for modeling (Table S8); TREEspot data visualization of KINOMEScan interactions of compound 12b, PQR620, and INK128 (Figure S6); selectivity profile calculated from KinomeScan data (Table S9); kinase interactions (KINOMEscan data) (Table S10); 1H NMR, 13C{1H} NMR, and NSI-HRMS spectra; HPLC chromatograms; chemical structures of final compounds and intermediates (PDF)
Compound 3a-PI3Kγ (PDB)
Compound 2a-mTOR (PDB)
Compound 2b-mTOR (PDB)
Compound 2a-PI3Kα (PDB)
Compound 2b-PI3Kα (PDB)
Molecular formula strings and some data (CSV)
PDB code 5JHB was used for docking of compound 3a into PI3Kγ. PDB code 4JT6 was used for docking of compounds 2a and 2b into mTOR kinase. PDB code 3ZIM was used for docking of compounds 2a and 2b into PI3Kα.
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.