Design, Synthesis, and Evaluation of Orally Bioavailable Quinoline

Feb 8, 2018 - A novel series of quinoline–indole derivatives were synthesized and evaluated as multitarget-directed ligands for the treatment of Alz...
3 downloads 6 Views 4MB Size
Subscriber access provided by UNIVERSITY OF TOLEDO LIBRARIES

Article

Design, Synthesis and Evaluation of Orally Bioavailable Quinoline-Indole Derivatives as Innovative MultitargetDirected Ligands: Promotion of Cell Proliferation in the Adult Murine Hippocampus for the Treatment of Alzheimer’s Disease Zhiren Wang, Jinhui Hu, Xiaoping Yang, Xing Feng, Xingshu Li, Ling Huang, and Albert S. C. Chan J. Med. Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jmedchem.7b01417 • Publication Date (Web): 08 Feb 2018 Downloaded from http://pubs.acs.org on February 8, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Journal of Medicinal Chemistry is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Design,

Synthesis

and

Evaluation

of

Orally

Bioavailable

Quinoline-Indole Derivatives as Innovative Multi-Target-Directed Ligands: Promotion of Cell Proliferation in the Adult Murine Hippocampus for the Treatment of Alzheimer’s Disease Zhiren Wang,†,‡ Jinhui Hu,† Xiaoping Yang,‡ Xing Feng,‡ Xingshu Li,*,† Ling Huang,*,† and Albert S. C. Chan† †

School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China



Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine,

Hunan Normal University, Changsha, 410013, China

Abstract A novel series of quinoline-indole derivatives were synthesized and evaluated as multi-target-directed ligands for the treatment of Alzheimer’s disease (AD). Biological evaluation revealed that the derivatives had multifunctional profiles including antioxidant effects, blood-brain barrier (BBB) penetration, biometal chelation, Aβ aggregation modulation and neurotrophic and neuroprotective properties. Moreover, several representative target derivatives demonstrated hippocampal cell proliferation in living adult mice by intracerebroventricular (ICV) injection or oral administration. Further drug-like property analysis demonstrated that the optimized compound, 8d (WI-1758), had liver microsomal metabolic stability, was well tolerated (>2000 mg/kg), and had a rational pharmacokinetic profile, as well as an oral bioavailability of 14.1% and a positive log BB (-0.19) to cross the BBB in vivo. Pharmacodynamics studies demonstrated that chronic oral administration of

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 2 of 91

8d•HCl substantially ameliorated the cognitive and spatial memory deficits in APP/PS1 AD mice and noticeably reduced overall cerebral β-amyloid deposits. Introduction Alzheimer’s disease (AD), the most prevalent and chronic type of neurodegenerative disease, affects approximately 47 million patients worldwide.1 The pathologic changes of AD are convoluted and multilayered; these changes include the aberrant accumulation of amyloid-β (Aβ) plaques, loss of synapses, neurofibrillary tangles within neurons, oxidative stress, dyshomeostasis of biometals in the brain, etc.2

Presently,

the

primary

therapeutic

options

for

AD

treatment

are

acetylcholinesterase inhibitors (AChEIs), such as donepezil, rivastigmine, and galantamine, as well as the N-methyl-D-aspartate (NMDA) receptor (NMDAR) antagonist, memantine, which can improve memory and cognitive function to a certain extent but cannot definitively cure this disease. Due to its complex pathogenesis, the multi-target-directed ligands (MTDLs) approach for AD treatment has been developed in both symptomatic and disease-modifying efficiencies3-6. Among the numerous neuropathological biomarkers of AD, the extracellular Aβ plaque deposits between neurons and intracellular neurofibrillary tangles within the brains of patient are primary markers.7 The self-assembled and hydrophobic aggregations of Aβ peptides are associated with complex pathways, soluble oligomers, paranuclei, and protofibrils, and they ultimately generate insoluble filaments.8 The dyshomeostasis of biometals is another biomarker, and through comparison with the age-matched healthy parenchyma within the brains of AD patients, 3-5-fold higher

ACS Paragon Plus Environment

Page 3 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

concentrations of metal ions in Aβ plaque deposits, such as Cu2+ (0.4 mM, 5.7 times), Zn2+ (1.0 mM, 2.8 times) and Fe2+/3+ (0.9 mM, 2.9 times), were found. These biometals have a comparatively greater affinity towards the N-terminus of the Aβ peptide, especially at residues His6, His13, and His14, which aggravated Aβ aggregation

and

exacerbated

their

noxious

properties.9

Furthermore,

the

dyshomeostasis redox active metal ions, including those unbonded and bonded with Aβ peptides, could exacerbate the overproduction of reactive oxygen species (ROS) that jeopardizes biological tissue and finally results in neuronal damage or apoptosis in the central nervous system (CNS) of AD patients.10 Consequently, the use of appropriate metal chelating agents to inhibit the production or accumulation of Aβ plaques and restore biometal homeostasis in the brains of AD patients has been developed as a potent therapeutic strategy for AD treatment.11 Clioquinol (CQ, Figure 1) and its analogs, with 8-hydroxy quinoline as a crucial scaffold, have a moderate affinity for metal ions and can extract metals from extracellular Aβ aggregates and shift them to copper-carrier proteins in the parenchymal cells without perturbing the overall essential metal biochemistry in the brain, demonstrating safety, noticeably strengthened cognition and substantially decreased cerebrospinal fluid (CSF) levels of Aβ in recent phase II clinical trials.12, 13 The more recently developed preclinical multi-target-directed

metal-chelating

agents

5-((methyl(prop-2-yn-1-yl)amino)methyl)quinolin-8-ol 11-(5-chloro-8-methoxyquinolin-2-yl)undecan-1-ol N1,N1-dimethyl-N4-(pyridin-2-ylmethyl)benzene-1,4-diamine

ACS Paragon Plus Environment

(M-30), (J2326), (L2-b)

and revealed

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 91

noticeable efficacies in multiple accessible murine models of AD (Figure 1).14-16

Figure 1. Structure of CQ, multi-target-directed metal-chelating agents, melatonin, melatonin-N-benzylamine

hybrids and several small molecules reported to manipulate neuronal cells in vitro and in vivo.

All adult mammalian species have reservoirs of neuronal stem cells in the hippocampal dentate gyrus (DG), which integrates new neurons into the hippocampus throughout life.17 This adult neurogenesis provides an indispensable mechanism by which lost or damaged neural cells from neurodegeneration can be replaced with functioning normal ones, which facilitates long-term hippocampal-dependent learning, memory and neuronal plasticity.18, 19 Nevertheless, with aging or neurodegeneration in rodents and elderly humans, the rate of regeneration of new neurons from the neural stem cells within the hippocampus decreases exponentially.20 Furthermore, before the new neural precursor cells are fully matured into functional neurons and migrate into the adult hippocampus, which is generally over approximately 30 days for mice, the vast majority of neural precursor cells undergo normal apoptosis during the first week with no explicit reason that has been currently identified.17,

18, 21

Therefore, the

promotion of adult hippocampal neurogenesis with small drug-like molecules was identified as an effective therapeutic strategy to address long-term neurodegeneration, which has significant unmet regenerative medical needs, particularly for AD.19

ACS Paragon Plus Environment

Page 5 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Allopregnanolone

(Figure

1)

and

the

compound

(6S,9aS)-N-benzyl-6-(4-hydroxybenzyl)-8-(naphthalen-1-ylmethyl)-4,7-dioxohexahyd ro-2H-pyrazino[1,2-a]pyrimidine-1(6H)-carboxamide

(ICG-001,

Figure

1)

significantly strengthened hippocampal neurogenesis and markedly reduced the pathological burden in a preclinical in vivo model of AD.22, 23 Through enhancing remyelination, benztropine (Figure 1) exhibited a striking mitigation of disease severity in a murine model of relapsing-remitting multiple sclerosis.24 Recently, the compound (P7C3A20,

N-(3-(3,6-dibromo-9H-carbazol-9-yl)-2-fluoropropyl)-3-methoxyaniline Figure

1)

and

its

further

analog

N-(3-(3,6-dibromo-9H-carbazol-9-yl)-2-fluoropropyl)-4-(4-(prop-2-yn-1-yloxy)benzo yl)benzamide (P7C3-S326, Figure 1), which has a preeminent ability to promote adult hippocampal neurogenesis, demonstrated significant efficacies in rodent models of Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), traumatic brain injury (TBI)

and

age-related

cognitive

decline.17,

18,

25-29

Interestingly,

the

melatonin-N-benzylamine hybrids (Figure 1), which have an indole framework, can promote the effective development of neural stem cells into the neuronal phenotype.6, 30

Taking into account neuronal loss, deficits in adult neurogenesis and the formation of plaques and tangles associated with AD, in this work, we merged the skeleton of indole and CQ to generate a series of quinoline-indole scaffolds to study their ability to modulate adult hippocampal neurogenesis in vivo, biometal dyshomeostasis and anti-amyloid aggregation (Figure 2). Through structure-activity

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

relationship (SAR) studies of antioxidant ability, BBB permeability, modulation of Aβ aggregation and neuronal cell proliferation activity, the potential compounds with neurotrophic function were further evaluated for their neuroprotective capacities, metabolic stability, acute toxicity and hippocampal cell proliferation in living adult mice. Ultimately, the pharmacokinetic profile and pharmacodynamics evaluation of the optimal compound was performed.

Figure 2. Multi-target-directed design strategy by introducing an indole fragment into the CQ framework.

Results and Discussion Chemistry The synthetic routes of the target compounds (7a-7g and 8a-8g) are depicted in Scheme 1. The commercially available 8-hydroxyquinaldine or one of its analogs (1a or 2a) was reacted with the corresponding o-nitrobenzaldehyde derivative to produce 3a-3g and 4a-4g, respectively, followed by reductive cyclization of the o-nitrostyrenes derivatives (3a-3g and 4a-4g) catalyzed by Pd(OAc)2 under CO, thus leading to the key indole intermediates 5a-5g and 6a-6g. Finally, the removal of the acetyl group by interesterification in the presence of CH3OH and K2CO3 (for 5a-5g) or NaOCH3 (for 6a-6g) led to the target compounds 7a-7g and 8a-8g with good yields. In addition, by treating the pure target compounds 7g, 8c, 8d and 8g with HCl (g) in an ethyl acetate solvent, the corresponding hydrochloride compounds were obtained with a high yield. The synthesis of the target compounds 9a, 9d and 10d is described

ACS Paragon Plus Environment

Page 6 of 91

Page 7 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

in Scheme 2. Compounds 2d and 2e, which were prepared according to a previous method,31 were reacted with chloro(methoxy)methane to give 2f and 2g. The subsequent reactions of 2f and 2g with o-nitrobenzaldehyde or its analogs in the presence of NaH gave 3i, 3j and 4j, respectively. Finally, target compounds 9a, 9d and 10d were produced by reductive cyclization of the o-nitrostyrene intermediates (3i, 3j or 4j) with CO, followed by deprotection of the methoxymethane groups under acidic conditions.

Scheme 1. Synthesis of 7a-7g (X = H), 8a-8g (X = Cl), 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl. Reagents and

conditions: a. acetic anhydride, 150 °C, 12 h; b. DMF, phen•H2O, Pd(OAc)2, CO, 80 °C, 24 h; c. CH3OH, K2CO3 (for 5a-5g) or NaOCH3 (for 6a-6g), rt, 1 h, d. AcOEt, HCl (g), rt, 12 h.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Scheme 2. Synthesis of 9a, 9d and 10d. Reagents and conditions: a. HCHO, HCl (g), rt, 72 h; b. P(OEt)3, 110 °C, 12 h; c. H2O, KOH, NaClO, rt, 2 h; d. CH2Cl2, DIPEA, ClMOM, rt, 5 h; e. DMF, NaH, rt, 4 h; f. DMF, phen•H2O, Pd(OAc)2, CO, 80 °C, 24 h; g. CH3OH, HCl (37%), rt, 5 h.

Free Oxygen Radical Scavenging Ability Oxidative stress is one of the earliest events in AD pathogenesis and oxidative damage is present within the brain of AD patients. Considering that 8-hydroxy quinoline derivatives generally have good metal ion complexing ability, we first determined the antioxidant ability of the quinoline-indole compounds, performed the free oxygen radical absorbance capacity (ORAC) assay using fluorescein (ORAC-FL) method with a vitamin E analog, Trolox, as the internal standard. Table 1 revealed that all of the target compounds exhibited significantly higher antioxidant efficacies compared with clioquinol (0.5), melatonin (2.4) and clioquinol + melatonin (2.9). The results also showed that the radical scavenging efficacies of the target compounds were principally derived from the indole moiety. An SAR study demonstrated that the phenolic hydroxyl groups on the indole moieties were pivotal for the antioxidant activity. Compounds 7f (5.4) and 8f (4.9), which have phenolic hydroxyl groups at the

ACS Paragon Plus Environment

Page 8 of 91

Page 9 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

5-position of the indole moieties, provided considerably better antioxidant efficacies compared with the analogs that were methylated of the hydroxyl group (7e, 3.5; 8e, 3.4). Notably, compound 9d (6.6), which has an indole moiety on the 5-position of the quinoline, demonstrated the greatest radical scavenging efficacy. The presence of the chlorine on the oxine framework indicated a slight detrimental effect on the antioxidant activity. Compounds 7d, 7f, and 9d, lacking a chlorine atom in the oxine scaffold, exhibited ORAC values of 5.9, 5.4 and 6.6, respectively, compared with 8d, 8f, and 10d (ORAC values 5.0, 4.9 and 5.3), which have a chlorine in the oxine scaffold. Table 1. Oxygen Radical Absorbance Capacity (ORAC, Trolox Equivalents), Permeability (Pe × 10-6 cm s-1) Determined by the PAMPA-BBB Assay for Target Compounds, Predicted Penetration of the CNS and Inhibition of

Aβ Self-aggregation.

Aβ1-42 self-induced Pe Comd.

R1

R2

X

ORACa

Pred. -6

aggregation

-1 b

(×10 cm s )

(%Inhib.)c

7a

H

H

H

4.5 ± 0.1

8.0 ± 1.2

CNS+

6.1 ± 1.3

7b

F

H

H

4.2 ± 0.3

10.6 ± 2.3

CNS+

34.2 ± 3.8

7c

Cl

H

H

4.6 ± 0.2

9.8 ± 1.6

CNS+

25.1 ± 3.9

7d

OH

H

H

5.9 ± 0.3

14.8 ± 0.7

CNS+

51.2 ± 9.1

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 91

7e

OMe

OMe

H

3.5 ± 0.2

5.2 ± 1.3

CNS+

21.5 ± 7.4

7f

OH

OMe

H

5.4 ± 0.1

12.1 ± 2.1

CNS+

42.3 ± 5.8

7g

-

-

H

4.3 ± 0.2

5.9 ± 0.3

CNS+

19.4 ± 5.0

8a

H

H

Cl

3.1 ± 0.2

6.6 ± 1.4

CNS+

13.6 ± 4.2

8b

F

H

Cl

3.2 ± 0.1

5.3 ± 0.4

CNS+

42.9 ± 5.1

8c

Cl

H

Cl

3.0 ± 0.1

5.6 ± 0.8

CNS+

35.9 ± 1.9

OH

H

Cl

5.0 ± 0.2

10.2 ± 0.1

CNS+

66.8 ± 8.2

8e

OMe

OMe

Cl

3.4 ± 0.2

3.9 ± 1.6

CNS±

32.8 ± 7.1

8f

OH

OMe

Cl

4.9 ± 0.1

8.4 ± 1.5

CNS+

50.3 ± 8.3

8g

-

-

Cl

3.2 ± 0.1

4.9 ± 0.2

CNS+

30.8 ± 5.6

9a

H

-

H

2.1 ± 0.1

12.8 ± 1.4

CNS+

43.1 ± 7.8

9d

OH

-

H

6.6 ± 0.1

9.1 ± 0.6

CNS+

57.2 ± 4.8

10d

OH

-

Cl

5.3 ± 0.2

6.8 ± 0.4

CNS+

68.7 ± 7.9

clioquinol

-

0.5 ± 0.2

-d

-d

1.9 ± 1.2

melatonin

-

2.4 ± 0.1

-d

-d

19.3 ± 3.4

-

2.9 ± 0.2

-d

-d

22.5 ± 5.3

curcumin

-

-d

-d

-d

36.7 ± 6.6

resveratrol

-

-d

-d

-d

42.1 ± 5.8

chlorpromazine

-

-d

5.9 ± 0.4

CNS+

-d

hydrocortisone

-

-d

1.1 ± 0.3

CNS–

-d

8d (WI-1758)

clioquinol + melatonin

ACS Paragon Plus Environment

Page 11 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

a

The means ± SD of three independent experiments. Data are expressed as µmol of trolox equivalent/µmol of

tested compound. b Compounds were dissolved in DMSO at 5 mg mL-1 and diluted with PBS/EtOH (70:30).

Values are expressed as the means ± SD at least three independent experiments, compounds with permeabilities values Pe > 4.7 × 10-6 cm s-1 could cross the blood-brain barrier by passive diffusion. c The Aβ1-42/inhibitor ratio was equal to 5/1, with inhibitor at 5 µM and Aβ1-42 at 25 µM. Values are the mean of three independent experiments in triplicate. d n.t. = not tested.

Blood-Brain Barrier Permeability Assay Penetration of the blood-brain barrier (BBB) is a critical factor for successful CNS drugs. To determine the BBB penetration of the target compounds, we used the parallel artificial membrane permeability (PAMPA) assay, which was initially established by Di et al and has been successfully applied to a multitude of diverse compounds to predict passive BBB permeation. The results presented in Table 1 indicated that most of the target compounds can significantly permeate the blood-brain barrier by passive diffusion. The SAR study indicated that compounds 7d (Pe = 14.8) and 7f (12.1), which contain a phenolic hydroxyl group on the indole moiety, demonstrated significantly higher BBB permeability compared with the analogs that replaced the phenolic hydroxyl groups with hydrogen (7a, 8.0), halogens (7b, 10.6; 7c, 9.8), dioxoles (7g, 5.9) or the methylation of the phenolic hydroxyl groups (7e, 5.2). In contrast, for compounds with the indole scaffold on the 5-position of the quinoline, such as 9a (12.8) and 9d (9.1), the phenolic hydroxyl groups on the indole moiety demonstrated slight unfavorable effects on BBB permeability. Likewise, the effect of chlorine on the oxine moiety displayed a slight undesirable effect on

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

brain penetration. The chloro-substituted compounds 8d (10.2) and 10d (6.8) exhibited slightly lower BBB permeabilities compared with the hydrogen-substituted counterparts 7d (14.8) and 9d (9.1). Inhibition of Aβ Self-Aggregation To determine the inhibitory activities of the quinoline-indole derivatives on Aβ1– 42

self-induced aggregation, the thioflavin T (ThT) fluorescence assay was first

performed with curcumin and resveratrol as reference compounds (Table 1). Among the four series of compounds, 7 (X = H), 8 (X = Cl), 9 (X = H) and 10 (X = Cl), compounds 7d (inhibition: 51.2%) and 8d (inhibition: 66.8%), in which a phenolic hydroxyl group was on the indole moiety at the R1 position, had more potent effects than clioquinol (1.9%), melatonin (19.3%), clioquinol + melatonin (22.5%) and the other analogs. Compounds 7f and 8f, with a methoxy group on the ortho (R2) position of the hydroxyl group, exhibited slightly weaker activities (42.3%, and 50.3%, respectively) than the parent compounds. The methylation of the hydroxyl group or cyclization with the methoxy group seemed unfavorable for the activities, and compounds 7e (21.5%), 8e (32.8%), 7g (19.4%) and 8g (30.8%) showed dramatic decreases in inhibiting Aβ1–42 aggregation, which demonstrated that the phenolic hydroxyl group on the indole moiety was preferable for the inhibitory activity. Replacement of the phenolic hydroxyl group with an H, F or Cl, as in compounds 7a-7c (6.1 – 34.2%), 8a-8c (13.6 – 42.9%), and 9a (43.1%), also provided much less potent activities than compounds 7d, 8d and 9d, respectively. Compounds 9d (57.2%) and 10d (68.7%), with the indole moiety on the 5-position of the quinoline, had

ACS Paragon Plus Environment

Page 12 of 91

Page 13 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

activities comparable with 7d and 8d. The results also demonstrated that the presence of a chlorine atom on the oxine moiety plays a pivotal role in the inhibitory activity, and compounds 8d (66.8%) and 10d (68.7%) exhibited more potent activities than their hydrogen-substituted counterparts 7d (51.2%) and 9d (57.2%), respectively. Metal-Chelating Property Studies Considering compound 8d showed excellent antioxidant ability, blood-brain barrier (BBB) permeability and inhibition of Aβ1–42 self-induced aggregation, and structurally, the quinoline-indole derivatives should also have good metal-chelating properties, we chose 8d as an typical for studying the metal-chelating ability of the quinoline-indole derivatives.31 The results in Figure 3 demonstrate that substantial new characteristic absorptions were detected at 338, 309, 324 and 309 nm after titrating 8d (314 nm) with CuSO4, ZnCl2, FeSO4 and FeCl3, respectively, which indicated that compound 8d was capable of chelating Cu2+, Zn2+, Fe2+, and Fe3+ and generating corresponding complexes. The stoichiometry of the Cu2+-8d complexes in buffer was initially determined using the inflexion point analysis, which measures the absorption at 338 nm by titrating 8d with Cu2+. As shown in Figure 3B, an isoabsorptive point (at 0.5) revealed a unique Cu2+-8d complex via a 1:2 Cu2+/ligand molar ratio. Moreover, high-resolution mass spectrometry was also performed to further probe the explicit components of the chelation complex, and the primary molecular ion peak of [(8d)2Cu]2+ (calculated: 375.0269, detected: 375.0277, Figure 3C) illustrated a molar ratio of 1:2 Cu2+/ligand for the complex.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 3. (A) UV absorbance spectrum of compound 8d (50 µM) alone or in the presence of CuSO4 (50 µM), ZnCl2 (50 µM) FeSO4 (50 µM) or FeCl3 (50 µM) in buffer (20 mM HEPES, 150 mM NaCl, pH = 7.4). (B) UV-vis titration of compound 8d (50 µM) with Cu2+ in buffer (20 mM HEPES, 150 mM NaCl, pH = 7.4) at room temperature. (C) High-resolution mass spectrometry of the Cu2+-8d complex in solution.

Inhibition of Self- and Metal-Induced Aβ Aggregation and Disaggregation of Aβ Aggregates by Quinoline-indole Derivatives To further elucidate the inhibitory effects of quinoline-indole derivatives on selfor metal-induced Aβ aggregation, translating to less toxic or non-toxic off-pathway amorphous Aβ aggregates, transmission electron microscopy (TEM) and the ThT assay were performed for both the inhibition of Aβ aggregates and disaggregation of Aβ aggregation fibrils in the presence of quinoline-indole derivatives. As indicated by the ThT assay (Figures 4), compounds 8d and 8g provided 71.6%-48.6% inhibition of self-induced Aβ1-42 aggregation and 72.7%-48.55% disaggregation of the preformed

ACS Paragon Plus Environment

Page 14 of 91

Page 15 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Aβ1-42 aggregate fibrils (Figures 4a and 4b), especially 8d, which were higher than the inhibition values of CQ (19.6% and 13.0%, respectively, p < 0.01 and 0.001) and melatonin (49.1% and 44.3%, respectively, p < 0.05 and 0.01). The morphological assessment of compound 8d by TEM (Figure 4c, sequence 5) showed less dense and thinner fibrils as well as fewer unstructured Aβ1-42 aggregates in the 8d-treated sample compared with the control (Figure 4c, sequence 2), CQ (Figure 4c, sequence 3) or melatonin (Figure 4c, sequence 4). Alternatively, in the disaggregation assay, 8d exhibited narrower and shorter aggregates (Figure 4d, sequence 5) than CQ (Figure 4d, sequence 3) and melatonin (Figure 4d, sequence 4). These results validated that 8d could effectively modulate self-induced Aβ1-42 aggregation by its overall structure rather than its structural components.

Figure 4. Experiments of inhibition self-induced Aβ1–42 aggregation and disaggregation of self-induced Aβ1–42 aggregates in the presence of 8d. (a and b) Top: Scheme for the inhibition or disaggregation experiments. Bottom:

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

The fluorescence intensity of the ThT binding assay, data are expressed as the mean ± SD at least three independent experiments. Statistical significance was analyzed by ANOVA: ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001. (c and d) TEM image analysis of the inhibition of Aβ1–42 aggregation and disaggregation of self-induced Aβ1–42 aggregates. Serial number: (1) fresh Aβ1–42, (2) Aβ1–42 alone, (3) Aβ1–42 + CQ, (4) Aβ1–42 + melatonin, (5) Aβ1–42 + 8d, (6) Aβ1–42 + 8g. Experimental conditions: Aβ1–42 (25 µM); compound/Aβ1-42 = 1/1; PBS (50 mM); pH 7.4; 37 °C.

The abilities of 8d to inhibit Cu2+-induced Aβ aggregation and induce the disaggregation of Aβ aggregation fibrils were investigated (Figure 5). The ThT fluorescence assay demonstrated that 8d had a rate of 85.8% inhibition for Aβ aggregation and 83.3% for disaggregation of Cu2+-associated Aβ aggregation fibrils, which were distinctly better than the values found for CQ (65.4% and 36.9%, respectively, p < 0.01 and 0.01) and melatonin (47.8% and 24.0%, respectively, p < 0.001 and 0.01). These results were further supported by TEM images of Aβ samples, which revealed the smaller-sized non-fibrillar amorphous Aβ species in the presence of Cu2+ and 8d (Figure 5c and 5d, sequence 5). In contrast, larger Aβ aggregates were observed with CQ (Figure 5c and 5d, sequence 4), melatonin (Figures 5c and 5d, sequence 6) and the control sample (Figures 5c and 5d, sequence 2). Because CQ exhibited a weaker ability to inhibit Cu2+-induced Aβ aggregation and to disaggregate the well-formed Aβ fibrils and melatonin provided almost no ability for either, these observations suggest that both the indole and 8-hydroxy quinoline moieties in 8d could be crucial for redirecting and interacting with metal-Aβ. The above results also demonstrated that substituents on the indole moiety of 8d, especially the hydroxyl

ACS Paragon Plus Environment

Page 16 of 91

Page 17 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

group, could influence the regulation of Aβ aggregation. These similar scenarios often appeared in the literature describing the development of specific chemical ligands for anti-amyloidogenic activity against metal-Aβ complexes.15, 32-34 Taken together, all of the results suggest that 8d has a strong ability to modulate self- and metal-induced Aβ aggregation.

Figure 5. Experiments of inhibition Cu2+-induced Aβ1–42 aggregation and disaggregation of Cu2+-induced Aβ1–42 aggregates in the presence of 8d. (a and b) Top: Scheme for the inhibition or disaggregation experiments. Bottom:

The fluorescence intensity of the ThT binding assay, data are expressed as the mean ± SD at least three independent experiments. Statistical significance was analyzed by ANOVA: ∗ p < 0.01, ∗∗ p < 0.01, ∗∗∗ p < 0.001. (c and d) TEM images analysis of the inhibition of Cu2+-induced Aβ1–42 aggregation and disaggregation of Cu2+-induced Aβ1–42 aggregates. Serial number: (1) fresh Aβ1–42, (2) Aβ1–42 alone, (3) Aβ1–42 + Cu2+, (4) Aβ1–42 + Cu2+ + CQ, (5) Aβ1–42 + Cu2+ + 8d, (6) Aβ1–42 + Cu2+ + melatonin. Experimental conditions: Aβ1–42 (25 µM); chelator/Aβ1-42/Cu2+ = 2/1/1; HEPES (20 µM) and NaCl (150 µM); pH 6.6; 37 °C.

Effects of the Quinoline-Indole Derivatives on Neuronal Proliferation of PC12

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Cells Inspired by the melatonin-N-benzylamine hybrids can promote the development of neural stem cells into the neuronal phenotype, we assessed the efficacies of the target compounds on neuronal proliferation with undifferentiated PC12 neuronal cells according to a well-established method.16, 35 After the cells were cultured for 72 h in the presence of the tested compounds (at concentrations ranging from 0.5 µM to 50 µM) or nerve growth factor (NGF, 50 ng/ml, as the positive control), the cells were surveyed under a digital microscope. As the results indicate in Figure 6A, compounds 7g, 8c, 8d and 8g did not show any neurotoxicity and revealed significant abilities to increase the number of cells. The SAR investigation indicated that replacement of the phenolic hydroxyl group on the indole scaffold of 8d (X = Cl, R1 = OH) with chlorine (8c, X = Cl, R1 = Cl) generated a slightly decreased neurotrophic activity. However, replacement with hydrogen (8a, X = Cl, R1 = H) and fluorine (8b, X = Cl, R1 = F) or the introduction of a methoxy group (8f, X = Cl, R1 = OH, R2 = OMe) resulted in the complete loss of the increased proliferation, and further methylation of the hydroxyl group (8e, X = Cl, R1 = R2 = OMe) displayed increased neurotoxicity. Similar trends were also observed for the compounds containing chlorine on the quinoline moiety, such as compounds 7c (X = H, R1 = Cl) and 7d (X = H, R1 = OH), and when the chlorine was removed from 8c or 8d, they exhibited a rather poor neurotrophic effect. In addition, shifting the indole moiety to the benzene ring of the quinoline, for example in compounds 9a, 9d and 10d, led to considerable neurotoxicity compared with the vehicle. Compounds 7g and 8g, which have a dioxole moiety on the indole,

ACS Paragon Plus Environment

Page 18 of 91

Page 19 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

demonstrated striking neurotrophic efficacies. The literature has reported that two mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinase (ERK) and p38, play crucial roles in neurotrophic actions.16, 36 Therefore, we used the ERK inhibitor U0126 (CAS: 109511-58-2) and the p38 inhibitor SB203580 (CAS: 152121-47-6) to study the effects of neuronal proliferation by quinoline-indole derivatives using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT, 570 nm) assay. With the addition of the ERK inhibitor U0126 (Figure 6B), the 8d-triggered proliferation of neuronal cells was completely abolished, and the addition of the p38 inhibitor SB203580 significantly decreased the proliferation effect of 8d. These preliminary results suggested that quinoline-indole derivatives might promote neuronal proliferation in a MAPK-dependent mechanism. Additionally, the morphological studies in Figure 7 shows that compounds 7g, 8c, 8d and 8g exhibited neurotrophic effects on PC12 cells, in contrast to the lack of increase in neurons using CQ and melatonin.

Figure 6. (A) The quantification of neuron number using image-based analysis. Statistics were expressed as the

percentage relative to control and as the mean ± SD of at least three independent experiments. NGF was tested at a

constant concentration of 50 ng/ml. (B) The effects of 8d, 8d + U0126 (ERK inhibitor, 1 µM) and 8d + SB203580

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(p38 inhibitor, 1 µM) on neuronal survival measured by MTT assay. Statistical significance was analyzed by ANOVA: ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 by compared with the control group (0 µM quinoline-indole derivatives).

Figure 7. Images from representative experiments showing the quinoline-indole derivatives in undifferentiated

PC12 cells: cells were treated with 50 ng/ml NGF or 10 µM of melatonin, CQ, 7g, 8c, 8d, or 8g, respectively.

Neuroprotective Capacity against Intracellular Oxidative Stress Cellular damage in the central nervous system by oxidative stress is a typical feature in AD, PD, Huntington’s disease (HD), and ALS, especially intracellular oxidative stress, such as mitochondrial ROS, which is closely involved in the most crucial intrinsic pathway leading to apoptosis emanating from the mitochondria.17 To explore the therapeutic potentials for compounds 7g, 8c, 8d and 8g as close as possible to the physiological conditions of the onset of neurodegenerative disease, an assay to investigate their neuroprotective capacities against intracellular oxidative stress was performed using the human neuroblastoma cell line SH-SY5Y (which is usually used in the determination of ROS) with tert-butyl hydroperoxide (t-BuOOH) as the ROS stimulant along with the fluorescence probe dichlorofluorescein diacetate (DCFH-DA), which was used to evaluate the intracellular ROS production. In the trial,

ACS Paragon Plus Environment

Page 20 of 91

Page 21 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

all compounds were tested at 1-0.01 µM, a concentration close to their concentrations for promoting neurite outgrowth. As shown in Figure 8, after t-BuOOH was introduced into the cells, the production of intracellular ROS units increased from 37.5 ± 5.2 (vehicle) to 415.5 ± 39.4 (control). In contrast, significantly less ROS production was detected in 7g-, 8c-, 8d- and 8g-incubated cells. The EC50 values (0.26, 0.82, 0.10 and 0.33 µM, respectively, for 7g, 8c, 8d and 8g) in Table 2 demonstrated that these compounds have the greatest capacities against intracellular oxidative stress compared with melatonin (EC50 > 1.0 µM) and the parent compound CQ (EC50 > 1.0 µM).

Figure 8. Inhibition of t-BuOOH-induced ROS production in the presence of CQ, melatonin, 7g, 8c, 8d and 8g

determined by DCFH-DA. Data are the mean ± SD of at least three independent experiments performed in

sextuplicate and expressed as fluorescence unit. Statistical comparisons with control (only t-BOOH) were

performed by one-way ANOVA. (levels of significance * p < 0.05;** p < 0.01;*** p < 0.001, ns p > 0.05).

Table 2. The EC50 Values of Neuroprotective Capacity against Intracellular Oxidative Stress in SH-SY5Y Cell.

Compound

EC50 (µM) a

Compound

ACS Paragon Plus Environment

EC50 (µM) a

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

a

Page 22 of 91

melatonin

> 1.0

clioquinol

> 1.0

7g

0.26 ± 0.07

8c

0.82 ± 0.12

8d

0.10 ± 0.03

8g

0.33 ± 0.05

Data are expressed as the mean ± SD at least three independent experiments.

Metabolic Stability in Liver Microsomes of SD Rats Proper liver microsomal stability of the optimal compounds is beneficial for the stability in metabolism. To evaluate this property, the metabolic stabilities of compounds 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl in the liver microsomes of SD rats were examined according the procedure described in a previous study with CQ and donepezil as the reference anti-AD agents and testosterone as a positive control.31 According to the results in Table 3, the hydrochlorides of 8c, 8d and 8g exhibited T1/2 values in liver microsomes of 195.8, 116.8 and 389.3 min, respectively, which demonstrated better stabilities compared with CQ (T1/2 = 34.4 min) and donepezil (T1/2 = 77.9 min). Compound 7g•HCl displayed a somewhat weaker stability and had a T1/2 of 58.6 min toward liver enzymes, which may be attributed to the lack of chlorine substituent to block the metabolic position of the quinoline scaffold. Overall, these results revealed that the hydrochlorides of 7g, 8c, 8d and 8g had considerable in vitro metabolic stabilities and thus deserve further detailed studies in vivo.37, 38 Table 3. Metabolic Stability of 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl in Liver Microsomes of SD Rats

Compound

k (min-1)

T1/2 (min) a

testosterone b

0.31323 ± 0.04206

2.2 ± 0.4

donepezil c

0.00889 ± 0.00041

77.9 ± 5.1

ACS Paragon Plus Environment

Page 23 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

a

CQ

0.02012 ± 0.00156

34.4 ± 3.8

7g•HCl

0.01183 ± 0.00026

58.6 ± 1.8

8c•HCl

0.00354 ± 0.00038

195.8 ± 23.1

8d•HCl

0.00593 ± 0.00020

116.8 ± 5.6

8g•HCl

0.00178 ± 0.00005

389.3 ± 15.5

Results are expressed as the mean ± SD of at least three independent experiments performed in triplicate. b, c The

positive control (testosterone) and the compound donepezil exhibited metabolic stability that was consistent with the literature and internal validation data.37, 38

Acute Toxicity Studies The acute toxicity profiles of the hydrochlorides of 7g, 8c, 8d and 8g were evaluated in adult C57BL/6 mice (12 weeks old). Each of the test compounds was given in single doses of 0, 677, 1333, or 2000 mg/kg by intragastric infusion (n = 6 for each dosage, three male and three female). After the administration of the compounds, any abnormal behavior and mortality changes were observed continuously during the first 4 h, intermittently for the following 24 h, and occasionally thereafter for 14 consecutive days for any delayed effects. During the observation period, no acute toxicity phenomena, including mortality, significant abnormal behaviors, drastically altered consumption of water or food and marked weight loss or gain, were observed. Additionally, no macroscopic abnormities of the stomach, intestines, heart, lung, liver, spleen, kidneys and other organs were detected after all of the mice were sacrificed on the 14th day. Furthermore, no significant histological abnormal changes were detected in the liver, heart, lung and brain of the

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

mice at the maximum tested dosage 2000 mg/kg in the histopathological studies by hematoxylin and eosin (HE) staining (Figure 9). Overall, these results revealed that 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl were well tolerated and exhibited no acute toxicity at a dose up to 2000 mg/kg.

Figure 9. Histological analysis of liver, heart, lung and brain for the acute toxicity studies of hydrochlorides of 7g,

8c, 8d and 8g at dosage of 2000 mg/kg in mice; 1000 µm and 100 µm indicate the scale bar of images (HE staining,

n = 6). Representative images of HE-stained liver, heart, lung and brain for each group are shown.

ACS Paragon Plus Environment

Page 24 of 91

Page 25 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Selective Promotion of Cell Proliferation in the Hippocampus of Living Adult Mice To further determine the efficacies of the target quinoline-indole compounds on cell proliferation in the hippocampus of living adult mice, an unbiased in vivo screening technique was performed.29 Each compound (hydrochlorides of 7g, 8c, 8d and 8g, at a concentration of 10 µM) was administered by intracerebroventricular (ICV) injection at a constant rate (0.5 µL/h) over 7 days into the left lateral ventricle of living adult wild type C57BL/6 mice (12 weeks old, n = 6 for each compound) via subcutaneously implanted osmotic minipumps.28 According to the previous studies, the attainable intracerebroventricular concentrations of the compounds were in a low-micromolar to mid-nanomolar concentration range.17 During the infusion of the compound, animals received a daily intraperitoneal (IP) injection with the thymidine analog bromodeoxyuridine (BrdU, 50 mg/kg/day) to mark new cells and the survival of proliferating hippocampal neural precursor cells. Apart from the ICV injection method, the administration of hydrochlorides of 7g, 8c, 8d and 8g (PO group, 30 mg/kg/day) by oral gavage to evaluate the efficacy of cell proliferation in the hippocampus of adult C57BL/6 mice was also performed (n = 6 for each group). To ensure a low baseline level of cell proliferation, mice were housed individually without access to running wheels seven days prior to the assay, as social interaction and voluntary exercise stimulate hippocampal neurogenesis. After one week of the administration of the compounds (including the ICV and PO groups), all animals were sacrificed and transcardially perfused. BrdU immunohistochemistry was used to

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

quantify cell proliferation by counting the number of BrdU+ cells in the brain hemisphere opposite to the side of infusion to avoid complications arising from the surgical implantation of the pump. The number of BrdU+ cells was normalized against the volume of the hippocampus to obtain consistent data across six animals.26 It is worth noting that the immunohistochemical staining of BrdU cells in the brains of mice illustrated that the quinoline-indole compounds could specifically incorporate BrdU in the hippocampal regions, including the DG, cornus ammonis (CA) 1, CA2 and CA3 regions, rather than the regions near the hippocampus, such as the cortex and striatum (Figure 10). This valid evidence further demonstrated that the target compounds had a preeminent ability to promote cell proliferation in the hippocampus of living adult mice rather than universally throughout the entire brain. This characteristic is very important for the treatment of AD, as all adult mammals form new neurons within the hippocampus, which is indispensable for learning, memory and neuronal plasticity.

Figure 10. The administration of quinoline-indole compounds by ICV indicated selective BrdU incorporation in

the hippocampus of living adult mice, including the dentate gyrus (DG), cornus ammonis (CA) 1, CA2 and CA3

regions, rather than cortex and striatum (immunohistochemical staining × 40, n = 6).

ACS Paragon Plus Environment

Page 26 of 91

Page 27 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

The cell proliferation efficacies displayed in Figures 11 and 12 demonstrated a substantial difference in the number of BrdU+ cells in the subgranular zone (SGZ) of DG regions between the quinoline-indole compound-treated mice, CQ- or melatonin-treated mice and the vehicle group. After the living mice were treated with the hydrochlorides of 7g, 8c, 8d and 8g over the 7 day period, the immunohistochemical results indicated that the rates of BrdU-incorporated new and surviving hippocampal neurons in the DG regions of the hippocampus were 43.6% (p < 0.001), 28.5% (p < 0.01), 41.3% (p < 0.001) and 31.2% (p < 0.01), respectively, which revealed strikingly greater results than for the vehicle (7.8%) and CQ (6.2%) groups. Melatonin displayed moderate effects (27.4%) on the hippocampal neurons, which was consistent with previously reported results.30 Consequently, the results indicated that the quinoline-indole compounds could promote cell proliferation from the reservoirs of neuronal stem cells in the subgranular zone (SGZ) of the hippocampal DG. Furthermore, the ICV injection of the hydrochlorides of 7g, 8c, 8d and 8g noticeably induced the cell proliferation of neural precursor cells in the CA1, CA2 and CA3 regions of the hippocampus (7g: 61.9%, 56.1% and 49.3%, p < 0.001; 8c: 15.3%, 18.1% and 20.3%, p < 0.01; 8d: 50.1%, 39.6% and 36.5%, p < 0.001; 8g: 49.8%, 38.0% and 37.6%, p < 0.001), which demonstrated significantly greater effectiveness compared with the vehicle (3.2%, 2.4% and 1.4%), CQ (2.7%, 2.1% and 1.5%) or melatonin (2.7%, 2.1% and 1.2%) groups (Figure 11 and 12). These results further suggested that compounds 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl can induce cell proliferation in the hippocampus of living adult mice.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 11. Immunohistochemically visualized BrdU incorporation in the new hippocampal neurons of living adult

C57BL/6 mice (immunohistochemical staining × 200, n = 6). The compounds 7g•HCl, 8c•HCl, 8d•HCl and

8g•HCl were administered ICV at the indicated concentration (0.5 µL/h) for 7 days, during which mice were dosed

IP daily with BrdU (50 mg/kg/day) to label new hippocampal neurons. CA1, CA2, CA3 and DG indicate the

cornus ammonis (CA) 1, CA2, CA3 and dentate gyrus (DG) regions of the hippocampus, respectively.

Representative 200 × magnification images of an immunohistochemically stained hippocampus for each group

were shown.

ACS Paragon Plus Environment

Page 28 of 91

Page 29 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Figure 12. Efficacy of 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl in the living adult mouse model of cell proliferation

within the hippocampus. The compounds were administered ICV at the indicated concentration (0.5 µL/h) for 7

days, during which time the mice were dosed IP daily with BrdU (50 mg/kg/day) to label new hippocampal

neurons. CA1, CA2, CA3 and DG indicate the cornus ammonis (CA) 1, CA2, CA3 and dentate gyrus (DG) regions

of the hippocampus, respectively. The results were expressed as the means ± SD (n = 6), and statistical significance was analyzed by two-way ANOVA: ns p > 0.05, ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 versus the vehicle, CQ and melatonin groups.

Alternatively, after oral gavage administration of compounds at the dose of 30 mg/kg/day for 7 days, the new hippocampal neurons in the DG, CA1, CA2 and CA3 regions of the hippocampus for quinoline-indole-compound treated groups (7g: 7.6%, 8.0%, 7.5% and 3.9%; 8c: 4.4%, 4.8%, 5.1% and 4.7%; 8d: 30.6%, 21.1%, 24.3% and 23.9%, p < 0.01; 8g:17.0%, 14.6%, 16.3% and 17.1%, p < 0.05) were higher than the vehicle (2.2%, 2.3%, 2.4% and 1.5%), CQ (2.0%, 2.4%, 2.3% and 1.4%) or melatonin

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(9.4%, 3.2%, 1.6% and 1.5%) groups (Figure 13 and 14). The reduced efficacies of compounds 7g•HCl and 8c•HCl by oral administration in contrast with ICV injection may be attributed their poor BBB permeabilities (See Table 1) and undesirable metabolic stabilities (See Table 2). Taken together, compound 8d•HCl exhibited a significant ability to selectively promote cell proliferation in the hippocampus by both ICV injection and oral administration, meeting the requirements of acceptable physicochemical properties for an oral drug in developing successful CNS drugs and drug candidates.39,

40

Therefore, compound 8d•HCl was chosen for further

pharmacokinetics investigation and pharmacodynamics evaluation.

Figure 13. Immunohistochemically visualized BrdU incorporation in the new hippocampal neurons of living adult

C57BL/6 mice (immunohistochemical staining × 200, n = 6). The compounds were administered orally at the

indicated dose (30 mg/kg/day) for 7 days, during which time mice were dosed IP daily with BrdU (50 mg/kg/day)

to label new hippocampal neurons. CA1, CA2, CA3 and DG indicate the cornus ammonis (CA) 1, CA2, CA3 and

ACS Paragon Plus Environment

Page 30 of 91

Page 31 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

dentate gyrus (DG) regions of the hippocampus, respectively. Representative 200 × magnification images of an

immunohistochemically stained hippocampus for each group were shown.

Figure 14. Efficacy of 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl in the adult mouse model of neurogenesis. The

compounds 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl were administered orally at the indicated dose (30 mg/kg/day)

for 7 days, during which time mice were dosed IP daily with BrdU (50 mg/kg/day) to label new hippocampal

neurons. CA1, CA2, CA3 and DG indicate the cornus ammonis (CA) 1, CA2, CA3 and dentate gyrus (DG) regions

of the hippocampus, respectively. The results were expressed as the means ± SD (n = 6), and statistical significance was analyzed by two-way ANOVA: ns p > 0.05, ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 versus the vehicle, CQ and melatonin groups.

Pharmacokinetic Profile and Log BB Values for the BBB Penetration of Compound 8d•HCl in SD Rats To evaluate the pharmacokinetic properties of 8d•HCl, a preliminary pharmacokinetics analysis in SD rats was performed. The results in Table 4

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 91

demonstrated that 8d•HCl had a C0 = 4926 µg/L, an AUC(0-∞) = 2124 h*µg/L, a moderate clearance of CL = 2.356 L/h/kg (0.6 L/h/kg < CL < 3.0 L/h/kg) and a temperate volume of distribution Vz = 2.491 L/kg (1 L/kg < Vz < 10 L/kg) for the intravenous administration (IV, dose = 5 mg/kg) mode. In addition, after single oral gavage administrations (PO, dose = 30 mg/kg), 8d•HCl had T1/2 = 3.58 h (T1/2 > 3.0 h), Tmax = 0.5 h (Tmax < 1.0 h), Cmax = 1001 µg/L and a rational oral bioavailability of F = 14.1%. Most importantly, the LC-MS/MS analysis of plasma and brain levels of 8d with the oral dose of 30 mg/kg revealed corresponding compound concentrations of 902 ng/g in the plasma and 579 ng/g in brain tissue after oral dosing for 1 h, which showed a log BB value of -0.19, which was substantially lower than the marginal value that determines whether a compound will be able to penetrate the BBB (log BB < -1). Overall, these favorable drug-like pharmacokinetic properties in vivo facilitated the pharmacodynamics evaluation of 8d•HCl in a murine model of AD. Table 4. Pharmacokinetic Profile of 8d•HCl in SD Rats.

AUC(0-t) IV

a

T1/2 (h)

C0 (µg/L) (h*µg/L)

(h*µg/L)

2124 ± 23

2491 ± 1724

AUC(0-t)

AUC(0-∞)

0.93 ± 0.21

4926 ± 266

2109 ± 14

PO b

T1/2 (h)

Tmax (h)

Cmax (µg/L)

3.58 ± 1.13

PO c (1h after dosing)

8d•HCl

0.5 ± 0.0

CL

MRT(0-t)

MRT(0-∞)

VSS

(mL/h/kg)

(h)

(h*µg/L)

(mL/kg)

2356 ± 24

0.38 ± 0.06

0.41 ± 0.07

954 ± 163

VZ (mL/kg)

8d•HCl

8d•HCl

AUC(0-∞)

1001 ± 128

(h*µg/L)

(h*µg/L)

1502 ± 49

1791 ± 323

MRT(0-t) (h)

MRT(0-∞) (h*µg/L)

F (%)

2.53 ± 0.74

4.88 ± 2.84

14.1

Plasma (ng/g)

Brain (ng/g)

Log BB (brain, log BB < -1)

902 ± 136

579 ± 89

-0.19 ± 0.06

ACS Paragon Plus Environment

Page 33 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

a

IV = intravenous administration, dose = 5 mg/kg, n = 3.

b

PO = oral administration, dose = 30 mg/kg, n = 3.

c

1 h after oral

administration, dose = 30 mg/kg, n = 3.

Cognitive and Memory Strengthening in APP/PS1 AD mice by Long-term Oral Uptake of 8d•HCl and 8g•HCl To evaluate the learning and memory efficacies of 8d•HCl and 8g•HCl in vivo, the Morris water maze (MWM) test, using double transgenic APP/PS1 mice (six months old), a well-established animal model of AD that exhibits early senile plaques in the brain and cognitive dysfunction, was performed (oral administration at 30 mg/kg/day; clioquinol, 30 mg/kg/day as the positive control) and wild type (WT) C57BL/6 mice were used as a negative control according to pharmacokinetic properties and previous studied dosage.13 The corresponding drug dosage was dissolved in a 0.5% carboxymethyl cellulose sodium (CMC-Na) solution by intragastric infusion for a total period of 80 days, and a blank 0.5% CMC-Na solution was used as a placebo for the WT and vehicle group mice. The results illustrated in Figure 15 indicated that during the entire administration period, 8d•HCl and 8g•HCl did not trigger any significant abnormalities in body weight compared with the vehicle- and CQ-treated mice, and additionally, no detrimental or abnormal events, including diarrhea, emesis-like behavior or an increased mortality were observed. During the final five days of treatment with the compounds, the Morris water maze (MWM) test of training trials was performed. The results indicated that the vehicle mice had adverse effects regarding spatial learning, and they spent more time and required more paths to locate the hidden escape platform in the pool compared with

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

the wild type mice (Figure 15, A2 and B). Conversely, the 8d•HCl- and 8g•HCl-treated groups, especially the 8d•HCl group (p < 0.01), had shortened mean escape latency and path searching in the APP/PS1 mice compared with the vehicle- or CQ-treated groups. This result revealed that the long-term oral intake of 8d•HCl significantly strengthened the spatial learning and memory capability in AD mice.

Figure 15. (A1) The mean daily body weight profile of each group mice (including APP/PS1 AD mice and WT

mice) during 80 consecutive days of oral drug administration period (WT = Wide Type mice, n = 10). (A2) The

escape latency time of each group was counted every day during the period of training trial (mean ± SD, n = 10). Statistical significance was analyzed by two-way ANOVA: ns p > 0.05, ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 versus the vehicle, compound 8d•HCl and 8g•HCl groups.. (B) The representative tracks of the mice in the Morris

water maze during the training trial period. The location of the platform and effective region (twofold diameter of

the platform) were represented as a blue and bright green circle, respectively.

Twenty-four hours after the last training, the long-term memory retention was assessed by performing the spatial probe trial. The platform in the pool was removed and the original platform location and its two-fold diameter location were set as the virtual platform and effective region, respectively. In the trial, each mouse was

ACS Paragon Plus Environment

Page 34 of 91

Page 35 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

permitted only one chance to swim freely for 90 seconds to search for the virtual platform. The results in Figure 16 (A1) indicated that the swimming velocities in the quadrants for groups treated with 8d•HCl, 8g•HCl, CQ and vehicle were approximately equivalent, which suggests that the animals’ locomotion and exploratory activities were not jeopardized by the long-term administration of compounds 8d•HCl and 8g•HCl (Figure 16, A1, p > 0.2, WT = 16.9 ± 4.5, vehicle = 17.8 ± 2.5, CQ = 18.7 ± 2.9, 8d•HCl = 18.9 ± 3.2 and 8g•HCl = 19.0 ± 1.9 cm/s). This effect further convincingly demonstrated the safety and non-toxic nature of 8d•HCl and 8g•HCl by long-term oral administration. The significantly higher numbers crossing the virtual platform (the original platform location, Figure 16, A2) and effective region (two-fold diameter of the original platform location, Figure 16, A3) in WT (4.8 ± 3.1, p < 0.001 and 5.7 ± 2.0, p < 0.001), 8d•HCl (4.5 ± 1.7, p < 0.001 and 4.9 ± 0.7, p < 0.001) and 8g•HCl (3.3 ± 2.4, p < 0.05 and 4.4 ± 0.5, p < 0.01) mice contrasted with their vehicle-treated (1.7 ± 1.1 and 2.1 ± 1.1) and CQ-treated (2.7 ± 1.6, p < 0.05 and 3.3 ± 1.9, p > 0.05) littermates and revealed the significantly better cognitive and memory function for 8d•HCl- and 8g•HCl-treated mice. The duration spent close to the platform location, including the time in the virtual platform (Figure 16, A4), effective region (Figure 16, A5) and correct quadrant (Figure 16, A6), for the 8d•HCl (3.2 ± 0.8, p < 0.05; 18.0 ± 8.3, p < 0.05 and 27.4 ± 5.1, p < 0.01) and 8g•HCl (2.6 ± 0.8, p > 0.05; 16.7 ± 8.0, p > 0.05 and 24.0 ± 4.1, p > 0.05) groups, especially the 8d•HCl group, were noticeably longer than for the vehicle (1.9 ± 0.2, 9.0 ± 3.8 and 20.6 ± 4.4) and CQ (2.1 ± 0.7, p > 0.05; 15.1 ± 5.3, p >

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

0.05 and 24.2 ± 3.9, p > 0.05) groups. These treatment outcomes further demonstrated that the oral uptake of 8d•HCl led to a considerable strengthening of the spatial memory retention and conventional cognitive capabilities compared with vehicle- or CQ-treated AD mice. Meanwhile, the swimming path length in the virtual platform (Figure 16, A7), effective region (Figure 16, A8) and correct quadrant (Figure 16, A9) for the 8d•HCl (22.1 ± 7.3, p < 0.001; 71.3 ± 17.1, p < 0.001 and 533.3 ± 71.1, p < 0.001) and 8g•HCl (15.0 ± 7.7, p < 0.05; 65.5 ± 16.9, p < 0.001 and 473.5 ± 27.2, p < 0.05) groups were remarkably longer than for the vehicle (9.1 ± 2.8; 44.2 ± 4.9 and 295.3 ± 46.9) and CQ (15.1 ± 5.3, p < 0.05; 45.8 ± 13.9, p > 0.05 and 416.4 ± 80.9, p < 0.001) groups, indicating that a more distinct specific spatial reference was formed in the memory vault for mice treated with compounds 8d•HCl and 8g•HCl. Taken together, these overall behavioral performance observations and outcomes demonstrated that the spatial memory impairment and cognitive dysfunction in vehicle-treated APP/PS1 mice were significantly ameliorated by the long-term oral administration of 8d•HCl compared with CQ at the comparative testing dosage.

ACS Paragon Plus Environment

Page 36 of 91

Page 37 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Figure 16. (A) Effect of long-term memory retention on the spatial probe trial (with the platform removed from

the pool) in the Morris water maze test after 80 consecutive days of oral drug administration treatment in APP/PS1

AD mice. The results were expressed as the means ± SD (n = 10). Statistical significance was analyzed by two-way ANOVA: ns p > 0.05, ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 versus the vehicle and compound 8d•HCl and 8g•HCl groups. (1) The average swimming speed for mice in each group. (2) Number of virtual platform (the

original platform location) crossings. (3) Number of effective region (two-fold diameter of the original platform

location) crossings. (4) The time spent in the virtual platform. (5) The time spent in the effective region. (6) The

time spent in the virtual platform quadrant (correct quadrant). (7) The swimming path length in the virtual platform.

(8) The swimming path length in the effective region (two-fold diameter of the platform). (9) The swimming path

length in the virtual platform quadrant (correct quadrant). (B) The representative tracks of the mice in the Morris

water maze during the spatial probe trial period. The location of the original platform and the effective region

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

(two-fold diameter of the platform) were represented as a blue and bright green circle, respectively.

Finally, all of the mice were sacrificed after the completion of the assays, and no expanded brain tissue or carcinogenicity in the subcutaneous layer, stomach, intestines, heart, lung, liver, spleen, kidneys and other organs or tissues were detected with 8d•HCl and 8g•HCl administration, which again suggested that 8d•HCl and 8g•HCl do not universally promote cell proliferation. In summary, these convincing results demonstrated that the long-term oral uptake of compounds 8d•HCl and 8g•HCl was well-tolerated and safe at doses of 30 mg/kg/day. Reduction of Amyloid Burden in APP/PS1 Transgenic Mice by Chronic Oral Administration of 8d•HCl and 8g•HCl The efficacy of 8d•HCl and 8g•HCl long-term oral treatment in strengthening learning and memory encouraged us to further investigate whether there was a correlation in the reduction of Aβ plaque deposition and quinoline-indole derivatives in APP/PS1 mice. The potential association of 8d•HCl and 8g•HCl with amyloid pathology was performed by Aβ plaque immunostaining in the brain tissue of APP/PS1 AD mice. The results revealed that 8d•HCl and 8g•HCl, especially 8d•HCl, treatment showed significant reductions in the amounts and area occupied by Aβ deposits in the coronal sections of the brain, including the cortex and hippocampus (Figure 17). Quantification analysis of the immunoreactivity revealed that 8d•HCl treatment demonstrated up to a 54% (p < 0.001, 8d•HCl vs. vehicle) reduction in plaque burden compared with vehicle littermate AD mice (Figure 18), which was substantially mitigated compared to CQ (35%, p < 0.05, 8d•HCl vs. CQ) treated AD

ACS Paragon Plus Environment

Page 38 of 91

Page 39 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

mice. In addition, a 26% decrease in the brain Aβ burden was detected after chronic treatment with 8g•HCl (p < 0.05, 8g•HCl vs. vehicle). In brief, these results suggest that chronic oral treatment with 8d•HCl and 8g•HCl markedly reduced the overall Aβ plaque deposition in APP/PS1 mice.

Figure 17. Oral drug administration of 8d•HCl and 8g•HCl reduces the total brain Aβ plaque burden in the

APP/PS1 transgenic mice after treatment at the dosage of 30 mg/kg/day for 80 consecutive days (n = 10).

Representative images of coronal sections from WT (wide type mice), vehicle, CQ-, 8d•HCl and 8g•HCl-treated

mice after immunohistochemical staining for Aβ with β-Amyloid (mOC64) antibody. Scale bar equals 1000 µm

and 400 µm, respectively.

Figure 18. The percentage of plaque burden in the overall half coronal hemisphere of mice was analyzed and

quantified by the area of Aβ immunoreactivity expressed as a percentage of full area using ImageJ software. The

results were expressed as the means ± SD (n = 10). Statistical significance was analyzed by two-way ANOVA: ns p > 0.05, ∗ p < 0.05, ∗∗ p < 0.01, ∗∗∗ p < 0.001 versus the vehicle and compound 8d•HCl and 8g•HCl groups.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Conclusion In summary, this work presented the design, synthesis and biological evaluation of a novel class of quinoline-indole derivatives as MTDLs against AD by merging the skeleton of indole and CQ. SAR studies revealed that most of the target derivatives had excellent free oxygen radical scavenging ability, BBB permeability by passive diffusion, inhibition of Aβ self-aggregation and stimulation of neuronal cell proliferation. The optimal compound, 8d•HCl (WI-1758), had a metal-chelating ability and an eminent neuroprotective capacity via intracellular antioxidant effects (EC50 = 0.1 µM), providing 71.6% and 85.8% inhibition of self- or Cu2+-induced Aβ aggregation as well as 72.7% and 83.3% disaggregation of preformed self- or Cu2+-associated Aβ1-42 aggregate fibrils. Moreover, 8d•HCl had reasonable pharmacokinetic properties, with substantial metabolic stability (T1/2 = 116.8 min in liver microsomes and T1/2 = 3.58 h in vivo), oral bioavailability (F = 14.1%), salient log BB values (log BB = -0.19) for penetration of the CNS in SD rats and no acute toxicity up to 2000 mg/kg. Further biological property analysis demonstrated that this optimal compound markedly and selectively promoted adult hippocampal cell proliferation in living adult C57BL/6 mice by ICV injection with osmotic minipumps or oral administration at a dosage of 30 mg/kg/day. Most importantly, pharmacodynamics studies by the Morris water maze test demonstrated that long-term (80 consecutive days) oral intake of compound 8d•HCl was not toxic and remarkably ameliorated cognitive and spatial memory deficits in the double transgenic APP/PS1 AD mice at a dosage of 30 mg/kg/day. Altogether, these convincing biochemical

ACS Paragon Plus Environment

Page 40 of 91

Page 41 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

results strongly highlight 8d•HCl as a reliable and potent drug candidate for treating AD and warrant further investigation of its mechanisms of action. EXPERIMENTAL SECTION Chemistry The NMR spectra were recorder using TMS as the internal standard on a Bruker AvanceIII spectrometer at 400.132 (1H NMR) and 100.614 (13C NMR) MHz. MS spectra were generated on an Agilent LC−MS 6120 instrument with an ESI and APCI mass selective detector. The melting points were determined using an SRS-Opti Melt automated melting point instrument. The reactions were followed by thin-layer chromatography (TLC) on glass-packed precoated silica gel plates and visualized in an iodine chamber or with a UV lamp. Flash column chromatography was performed using silica gel (200–300 mesh) purchased from Qingdao Haiyang Chemical Co. Ltd. The high-resolution mass spectra were obtained using a Shimadzu LCMS-IT-TOF mass spectrometer. The purity ( ≥ 95%) of the samples was determined by HPLC, conducted on a Shimadzu LC-20AT series system, TC-C18 column (4.6×250 mm, 5 µm), eluted with CH3CN/H2O = 90/10, at a flow rate of 0.5 mL/min.

5-(Chloromethyl)quinolin-8-ol hydrochloride (2c•HCl) Compound 2c•HCl were synthesised according to the literature.41 Pale yellow solid, 98% yield. Rf = 0.42 (petroleum/EtOAc = 1/1). 1H NMR (400 MHz, DMSO) δ 9.32 (t, J = 7.7 Hz, 1H), 9.14 (dd, J = 19.3, 4.9 Hz, 1H), 8.16 (ddd, J = 36.0, 8.6, 5.3 Hz, 1H),

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

7.80 (dd, J = 75.6, 8.0 Hz, 1H), 7.57 (t, J = 7.4 Hz, 1H), 5.32 (s, 1H), 4.90 (s, 1H). LC/MS (ESI): 194.0 [M+H]+. Diethyl ((8-hydroxyquinolin-5-yl)methyl)phosphonate (2d)31

To a solution of triethyl phosphite (18.86 mL, 3.0 equiv) was added 1.0 equiv of compound 2c (7.0 g) in one portion. The mixture was stirred at 110 °C for 12 h. After the completion of the reaction, the residue was directly purified by flash chromatography on silica gel with ethyl acetate as the elution solvent to afford the pure product. Pale yellow solid, 78% yield. Rf = 0.32 (CH2Cl2/CH3OH = 20/1). 1H NMR (400 MHz, CDCl3) δ 8.79 (dd, J = 4.2, 1.3 Hz, 1H), 8.45 (dd, J = 8.6, 1.4 Hz, 1H), 7.49 (dd, J = 8.6, 4.2 Hz, 1H), 7.40 (dd, J = 7.9, 3.8 Hz, 1H), 7.13 (dd, J = 7.9, 0.9 Hz, 1H), 4.03 – 3.88 (m, 4H), 3.50 (d, J = 21.3 Hz, 2H), 1.18 (t, J = 7.1 Hz, 6H). LC/MS (ESI): 296.1 [M+H]+.

Diethyl ((7-chloro-8-hydroxyquinolin-5-yl)methyl)phosphonate (2e)

Compound 2d (5.0 g) was suspended in 30 mL of H2O containing 17 mL of 1 N KOH solution. 15.3 mL of NaClO solution (7.5 % excess) was added in 30 min with vigorously stirring, and then stirred at room temperature. After the completion of the reaction (monitored by TLC), the solution was adjusted to pH 6.0 by adding 10 % hydrochloric acid, and then filtrated to get the solid product without further purification. White solid, 84% yield. Rf = 0.33 (CH2Cl2/CH3OH = 20/1).1H NMR (400 MHz, CDCl3) δ 8.81 (dd, J = 4.1, 1.0 Hz, 1H), 8.44 (dd, J = 8.6, 1.2 Hz, 1H),

ACS Paragon Plus Environment

Page 42 of 91

Page 43 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

7.51 (dd, J = 8.6, 4.2 Hz, 1H), 7.45 (d, J = 3.8 Hz, 1H), 4.05 – 3.93 (m, 4H), 3.50 (s, 1H), 3.44 (s, 1H), 1.21 (t, J = 7.1 Hz, 6H). LC/MS (ESI): 330.1 [M+H]+.

General Procedures for the Synthesis of 2f-2g A mixture of 2d or 2e (4.0 g, 1.0 equiv) and diisopropylethylamine (4.0 equiv) in 20 mL of DCM was added 1.5 equiv of chloromethyl methyl ether in dropwise, the mixture was stirred at room temperature. After the completion of the reaction, the solvent was evaporated under vacuum, and the crude product was purified by silica-column chromatography. Diethyl ((8-(methoxymethoxy)quinolin-5-yl)methyl)phosphonate (2f) Yellow oil. 72% yield. Rf = 0.32 (CH2Cl2/CH3OH = 20/1). 1H NMR (400 MHz, CDCl3) δ 8.96 (dd, J = 4.1, 1.4 Hz, 1H), 8.43 (dd, J = 8.6, 1.5 Hz, 1H), 7.48 (dd, J = 8.6, 4.1 Hz, 1H), 7.42 (dd, J = 8.1, 3.6 Hz, 1H), 7.38 (d, J = 8.2 Hz, 1H), 5.50 (s, 2H), 4.03 – 3.89 (m, 4H), 3.57 (s, 3H), 3.55 (s, 1H), 3.49 (s, 1H), 1.17 (t, J = 7.1 Hz, 6H). LC/MS (ESI): 340.1 [M+H]+. Diethyl ((7-chloro-8-(methoxymethoxy)quinolin-5-yl)methyl)phosphonate (2g) Yellow oil. 75% yield. Rf = 0.36 (CH2Cl2/CH3OH = 20/1). 1H NMR (400 MHz, CDCl3) δ 8.92 (dd, J = 4.0, 1.3 Hz, 1H), 8.42 (dd, J = 8.6, 1.5 Hz, 1H), 7.50 (d, J = 3.8 Hz, 1H), 7.46 (dd, J = 8.6, 4.1 Hz, 1H), 5.62 (s, 2H), 4.05 – 3.95 (m, 4H), 3.72 (s, 3H), 3.53 (s, 1H), 3.48 (s, 1H), 1.21 (t, J = 7.1 Hz, 6H). LC/MS (ESI): 374.0 [M+H]+. General Procedures for the Synthesis of 3a-3g and 4a-4g Quinoline (1.0 equiv, 1.0 g) and benzaldehyde (1.0 equiv) was dissolved in 20 mL of acetic anhydride, the mixture was stirred at 150 °C for about eight hours. After

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

cooled to room temperature, the mixture was filtrated, washed with water and dried under vacuum to get the product. (E)-2-(2-Nitrostyryl)quinolin-8-yl acetate (3a) Yellow solid. 82% yield. Rf = 0.29 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.24 (d, J = 16.0 Hz, 1H), 8.18 (d, J = 8.6 Hz, 1H), 8.01 (dd, J = 8.2, 1.1 Hz, 1H), 7.88 (d, J = 7.9 Hz, 1H), 7.70 (dd, J = 8.1, 1.3 Hz, 1H), 7.64 (dd, J = 10.2, 4.5 Hz, 2H), 7.49 (dt, J = 7.1, 5.5 Hz, 2H), 7.44 (dd, J = 7.5, 1.3 Hz, 1H), 7.33 (d, J = 16.0 Hz, 1H), 2.57 (s, 3H). LC/MS (ESI): 335.1 [M+H]+. (E)-2-(5-Fluoro-2-nitrostyryl)quinolin-8-yl acetate (3b) Yellow solid. 75% yield. Rf = 0.31 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 8.28 (dd, J = 15.9, 1.0 Hz, 1H), 8.19 (d, J = 8.5 Hz, 1H), 8.10 (dd, J = 9.1, 5.1 Hz, 1H), 7.71 (dd, J = 8.1, 1.3 Hz, 1H), 7.65 (d, J = 8.5 Hz, 1H), 7.55 (dd, J = 9.7, 2.3 Hz, 1H), 7.51 (d, J = 8.0 Hz, 1H), 7.45 (dd, J = 7.5, 1.4 Hz, 1H), 7.31 (d, J = 16.0 Hz, 1H), 7.18 – 7.12 (m, 1H), 2.57 (s, 3H).LC/MS (ESI): 353.1 [M+H]+. (E)-2-(5-Chloro-2-nitrostyryl)quinolin-8-yl acetate (3c) Yellow solid. 89% yield. Rf = 0.33 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.22 (d, J = 16.0 Hz, 1H), 8.15 (d, J = 8.5 Hz, 1H), 7.97 (d, J = 8.7 Hz, 1H), 7.83 (d, J = 2.1 Hz, 1H), 7.67 (dd, J = 8.0, 1.3 Hz, 1H), 7.61 (d, J = 8.5 Hz, 1H), 7.50 (t, J = 7.8 Hz, 1H), 7.44 (dd, J = 7.5, 1.4 Hz, 1H), 7.41 (dd, J = 8.7, 2.2 Hz, 1H), 7.29 (d, J = 15.9 Hz, 1H), 2.57 (s, 3H). LC/MS (ESI): 369.0 [M+H]+. (E)-2-(5-Acetoxy-2-nitrostyryl)quinolin-8-yl acetate (3d)

ACS Paragon Plus Environment

Page 44 of 91

Page 45 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Yellow solid. 81% yield. Rf = 0.36 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 8.27 (d, J = 16.0 Hz, 1H), 8.16 (d, J = 8.5 Hz, 1H), 8.08 (d, J = 8.9 Hz, 1H), 7.68 (dd, J = 8.0, 1.1 Hz, 1H), 7.62 (d, J = 8.6 Hz, 1H), 7.60 (d, J = 2.4 Hz, 1H), 7.50 (t, J = 7.8 Hz, 1H), 7.44 (dd, J = 7.4, 1.2 Hz, 1H), 7.28 (d, J = 16.0 Hz, 1H), 7.22 (dd, J = 8.9, 2.4 Hz, 1H), 2.57 (s, 3H), 2.37 (s, 3H). LC/MS (ESI): 393.1 [M+H]+. (E)-2-(4,5-Dimethoxy-2-nitrostyryl)quinolin-8-yl acetate (3e) Yellow solid. 92% yield. Rf = 0.26 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.50 (d, J = 8.8 Hz, 1H), 8.42 (d, J = 16.0 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 7.65 (d, J = 8.3 Hz, 2H), 7.20 (t, J = 8.0 Hz, 2H), 4.07 (s, 3H), 4.00 (s, 3H), 2.60 (s, 3H). LC/MS (ESI): 395.1 [M+H]+. (E)-2-(5-Acetoxy-4-methoxy-2-nitrostyryl)quinolin-8-yl acetate (3f) Yellow solid. 72% yield. Rf = 0.25 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 8.26 (dd, J = 15.9, 3.9 Hz, 1H), 8.17 (dd, J = 8.4, 4.2 Hz, 1H), 7.70 (dd, J = 7.1, 3.5 Hz, 1H), 7.67 – 7.60 (m, 2H), 7.58 (d, J = 4.0 Hz, 1H), 7.54 – 7.47 (m, 1H), 7.47 – 7.41 (m, 1H), 7.23 (dd, J = 16.1, 4.2 Hz, 1H), 3.96 (s, 3H), 2.58 (s, 3H), 2.39 (s, 3H). LC/MS (ESI): 423.1 [M+H]+. (E)-2-(2-(6-Nitrobenzo[d][1,3]dioxol-5-yl)vinyl)quinolin-8-yl acetate (3g) Yellow solid. 89% yield. Rf = 0.32 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 8.28 (d, J = 15.8 Hz, 1H), 8.16 (d, J = 8.2 Hz, 1H), 7.66 (dd, J = 20.5, 8.1 Hz, 2H), 7.55 – 7.42 (m, 3H), 7.25 – 7.15 (m, 2H), 6.15 (s, 2H), 2.56 (s, 3H). LC/MS (ESI): 379.1 [M+H]+. General Procedures for the Synthesis of 3i-3j and 4j

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

To a solution of 2f or 2g (1.0 equiv) in 10 mL anhydrous DMF, 3.0 equiv of sodium hydride (80 %) was added in portion at ice bath. The mixture was stirred for 30 min, a solution of the corresponding aldehyde (1.0 equiv) in 2 mL of DMF was added in dropwise and the reaction was monitored by TLC. After the completion of the reaction, water was added and the product was extracted by DCM, washed with brine, dried over anhydrous Na2SO4, evaporated the solvent under reduced pressure to afford the crude product which was purified by silica-column chromatography. (E)-8-(Methoxymethoxy)-5-(2-nitrostyryl)quinoline (3i) Yellow solid. 70% yield. Rf = 0.32 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.00 (dd, J = 4.1, 1.6 Hz, 1H), 8.54 (dd, J = 8.6, 1.6 Hz, 1H), 8.01 (dd, J = 8.2, 1.1 Hz, 1H), 7.84 (d, J = 7.8 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.70 – 7.62 (m, 2H), 7.58 (d, J = 15.8 Hz, 1H), 7.51 (dd, J = 8.6, 4.1 Hz, 1H), 7.49 – 7.41 (m, 2H), 5.55 (s, 2H), 3.60 (s, 3H). LC/MS (ESI): 337.1 [M+H]+. (E)-8-(Methoxymethoxy)-5-(5-(methoxymethoxy)-2-nitrostyryl)quinoline (3j) Yellow solid. 91% yield. Rf = 0.28 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 9.00 (dd, J = 4.1, 1.5 Hz, 1H), 8.54 (dd, J = 8.6, 1.5 Hz, 1H), 8.10 (d, J = 9.1 Hz, 1H), 7.79 (d, J = 8.2 Hz, 1H), 7.69 (d, J = 15.8 Hz, 1H), 7.60 (d, J = 15.8 Hz, 1H), 7.51 (dd, J = 8.6, 4.1 Hz, 1H), 7.47 (d, J = 8.2 Hz, 1H), 7.38 (d, J = 2.6 Hz, 1H), 7.12 – 7.02 (m, 1H), 5.55 (s, 2H), 5.32 (s, 2H), 3.60 (s, 3H), 3.54 (s, 3H). LC/MS (ESI): 397.1 [M+H]+ (E)-5,7-Dichloro-2-(2-nitrostyryl)quinolin-8-yl acetate (4a)

ACS Paragon Plus Environment

Page 46 of 91

Page 47 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Yellow solid. 91% yield. Rf = 0.28 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 8.49 (d, J = 8.7 Hz, 1H), 8.30 (d, J = 15.9 Hz, 1H), 8.02 (d, J = 8.1 Hz, 1H), 7.86 (d, J = 7.6 Hz, 1H), 7.76 – 7.58 (m, 3H), 7.50 (t, J = 7.6 Hz, 1H), 7.29 (d, J = 17.1 Hz, 1H), 2.60 (s, 3H). LC/MS (ESI): 403.0 [M+H]+. (E)-5,7-Dichloro-2-(5-fluoro-2-nitrostyryl)quinolin-8-yl acetate (4b) Yellow solid. 87% yield. Rf = 0.23 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.51 (d, J = 8.7 Hz, 1H), 8.33 (dd, J = 15.9, 0.9 Hz, 1H), 8.11 (dd, J = 9.1, 5.1 Hz, 1H), 7.70 (d, J = 8.8 Hz, 1H), 7.65 (s, 1H), 7.54 – 7.50 (m, 1H), 7.27 (d, J = 15.9 Hz, 1H), 7.20 – 7.14 (m, 1H), 2.60 (s, 3H). LC/MS (ESI): 421.0 [M+H]+. (E)-5,7-Dichloro-2-(5-chloro-2-nitrostyryl)quinolin-8-yl acetate (4c) Yellow solid. 79% yield. Rf = 0.27 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.48 (d, J = 8.7 Hz, 1H), 8.27 (d, J = 15.9 Hz, 1H), 8.00 (d, J = 8.8 Hz, 1H), 7.81 (d, J = 2.2 Hz, 1H), 7.67 (d, J = 8.8 Hz, 1H), 7.64 (s, 1H), 7.44 (dd, J = 8.7, 2.2 Hz, 1H), 7.26 (d, J = 15.9 Hz, 1H), 2.61 (s, 3H). LC/MS (ESI): 437.0 [M+H]+. (E)-2-(5-Acetoxy-2-nitrostyryl)-5,7-dichloroquinolin-8-yl acetate (4d) Yellow solid. 92% yield. Rf = 0.39 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.50 (d, J = 8.8 Hz, 1H), 8.33 (d, J = 15.9 Hz, 1H), 8.10 (d, J = 8.9 Hz, 1H), 7.69 (d, J = 8.8 Hz, 1H), 7.64 (s, 1H), 7.59 (d, J = 2.4 Hz, 1H), 7.27 (d, J = 9.7 Hz, 1H), 7.23 (d, J = 2.6 Hz, 1H), 2.60 (s, 3H), 2.38 (s, 3H). LC/MS (ESI): 461.0 [M+H]+. (E)-5,7-Dichloro-2-(4,5-dimethoxy-2-nitrostyryl)quinolin-8-yl acetate (4e) Yellow solid. 87% yield. Rf = 0.33 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.50 (d, J = 8.8 Hz, 1H), 8.42 (d, J = 16.0 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H),

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

7.65 (d, J = 8.3 Hz, 2H), 7.20 (t, J = 8.0 Hz, 2H), 4.07 (s, 3H), 4.00 (s, 3H), 2.60 (s, 3H). LC/MS (ESI): 463.0 [M+H]+. (E)-2-(5-Acetoxy-4-methoxy-2-nitrostyryl)-5,7-dichloroquinolin-8-yl acetate (4f) Yellow solid. 68% yield. Rf = 0.38 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.48 (d, J = 8.7 Hz, 1H), 8.30 (d, J = 15.9 Hz, 1H), 7.84 – 7.60 (m, 3H), 7.55 (s, 1H), 7.26 (s, 1H), 7.19 (d, J = 15.9 Hz, 1H), 3.96 (s, 3H), 2.61 (s, 3H), 2.39 (s, 3H). LC/MS (ESI): 491.0 [M+H]+. (E)-5,7-Dichloro-2-(2-(6-nitrobenzo[d][1,3]dioxol-5-yl)vinyl)quinolin-8-yl acetate (4g) Yellow solid. 72% yield. Rf = 0.31 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.44 (d, J = 8.7 Hz, 1H), 8.31 (d, J = 16.0 Hz, 1H), 7.71 – 7.57 (m, 2H), 7.53 (s, 1H), 7.26 – 6.93 (m, 2H), 6.16 (s, 2H), 2.60 (s, 3H). LC/MS (ESI): 447.0 [M+H]+. (E)-7-Chloro-8-(methoxymethoxy)-5-(5-(methoxymethoxy)-2-nitrostyryl)quinolin e (4j) Yellow solid. 70% yield. Rf = 0.31 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.95 (dd, J = 4.1, 1.6 Hz, 1H), 8.50 (dd, J = 8.6, 1.6 Hz, 1H), 8.12 (d, J = 9.1 Hz, 1H), 7.83 (s, 1H), 7.69 (d, J = 15.8 Hz, 1H), 7.51 (d, J = 16.7 Hz, 1H), 7.47 (dd, J = 8.7, 4.2 Hz, 1H), 7.35 (d, J = 2.6 Hz, 1H), 7.10 (dd, J = 9.1, 2.6 Hz, 1H), 5.67 (s, 2H), 5.32 (s, 2H), 3.74 (s, 3H), 3.54 (s, 3H). LC/MS (ESI): 431.1 [M+H]+. General Procedures for the Synthesis of 5a-5g, 5i-5j, 6a-6g and 6j.

ACS Paragon Plus Environment

Page 48 of 91

Page 49 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

An autoclave was charged with Pd(OAc)2 (0.05 equiv), 1,10-phenathroline•H2O (0.1 equiv) and 5 mL DMF. After the mixture was stirred at room temperature for 30min, 1.0 g of nitro-compound (1.0 equiv) was added and the vessel was purged six times with CO, and then pressurized to 40 bar with this gas. After the mixture was stirred at 80 °C for overnight, 100 mL of water was added, then, filtrated, washed with pure water and dried under vacuum to afford the product.

2-(1H-Indol-2-yl)quinolin-8-yl acetate (5a) Yellow solid. 88% yield. Rf = 0.23 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.46 (s, 1H), 8.19 (d, J = 8.6 Hz, 1H), 7.94 (d, J = 8.7 Hz, 1H), 7.71 (dd, J = 7.6, 2.0 Hz, 2H), 7.57 – 7.40 (m, 3H), 7.31 – 7.27 (m, 1H), 7.20 (s, 1H), 7.16 (t, J = 7.5 Hz, 1H), 2.62 (s, 3H). LC/MS (ESI): 303.3 [M+H]+. 2-(5-Fluoro-1H-indol-2-yl)quinolin-8-yl acetate (5b) Yellow solid. 65% yield. Rf = 0.34 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.44 (s, 1H), 8.18 (d, J = 8.6 Hz, 1H), 7.89 (d, J = 8.6 Hz, 1H), 7.70 (d, J = 7.7 Hz, 1H), 7.58 – 7.40 (m, 2H), 7.32 (dt, J = 9.8, 5.1 Hz, 2H), 7.11 (s, 1H), 7.01 (td, J = 9.1, 2.3 Hz, 1H), 2.59 (s, 3H). LC/MS (ESI): 321.1 [M+H]+. 2-(5-Chloro-1H-indol-2-yl)quinolin-8-yl acetate (5c) Yellow solid. 74% yield. Rf = 0.58 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.46 (s, 1H), 8.19 (d, J = 8.7 Hz, 1H), 7.90 (d, J = 8.6 Hz, 1H), 7.70 (dd, J = 7.9, 1.5 Hz, 1H), 7.64 (d, J = 1.9 Hz, 1H), 7.50 (t, J = 7.7 Hz, 1H), 7.45 (dd, J = 7.5, 1.6 Hz, 1H), 7.35 (d, J = 8.6 Hz, 1H), 7.21 (dd, J = 8.7, 2.0 Hz, 1H), 7.09 (d, J = 1.2 Hz, 1H), 2.58 (s, 3H). LC/MS (ESI): 337.1 [M+H]+.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

2-(5-Acetoxy-1H-indol-2-yl)quinolin-8-yl acetate (5d) Yellow solid. 83% yield. Rf = 0.21 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.45 (s, 1H), 8.15 (d, J = 8.7 Hz, 1H), 7.88 (d, J = 8.7 Hz, 1H), 7.68 (dd, J = 7.7, 1.8 Hz, 1H), 7.50 – 7.42 (m, 2H), 7.36 (dd, J = 5.5, 3.3 Hz, 2H), 7.12 (d, J = 1.3 Hz, 1H), 6.97 (dd, J = 8.8, 2.2 Hz, 1H), 2.58 (s, 3H), 2.33 (s, 3H). LC/MS (ESI): 361.1 [M+H]+. 2-(5,6-Dimethoxy-1H-indol-2-yl)quinolin-8-yl acetate (5e) Yellow solid. 61% yield. Rf = 0.35 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 9.48 (s, 1H), 8.11 (d, J = 8.7 Hz, 1H), 7.84 (d, J = 8.7 Hz, 1H), 7.66 (dd, J = 6.7, 2.8 Hz, 1H), 7.54 – 7.29 (m, 2H), 7.06 (s, 1H), 7.05 (d, J = 1.8 Hz, 1H), 6.75 (s, 1H), 3.93 (s, 3H), 3.88 (s, 3H), 2.55 (s, 3H). LC/MS (ESI): 363.1 [M+H]+. 2-(5-Acetoxy-6-methoxy-1H-indol-2-yl)quinolin-8-yl acetate (5f) Yellow solid. 79% yield. Rf = 0.22 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, DMSO) δ 8.41 (d, J = 8.7 Hz, 1H), 8.12 (d, J = 8.7 Hz, 1H), 7.85 (d, J = 7.3 Hz, 1H), 7.54 (q, J = 7.4 Hz, 2H), 7.32 (s, 2H), 7.20 (s, 1H), 3.84 (s, 3H), 2.60 (s, 3H), 2.28 (s, 3H). LC/MS (ESI): 391.2 [M+H]+. 2-(8H-[1,3]Dioxolo[4,5-g]indol-7-yl)quinolin-8-yl acetate (5g) Yellow solid. 81% yield. Rf = 0.25 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.34 (s, 1H), 8.10 (d, J = 8.7 Hz, 1H), 7.80 (dd, J = 8.7, 4.7 Hz, 1H), 7.65 (dd, J = 6.7, 2.8 Hz, 1H), 7.44 – 7.42 (m, 1H), 7.28 (dd, J = 8.3, 1.0 Hz, 1H), 7.05 – 6.99 (m, 2H), 6.86 (d, J = 14.5 Hz, 1H), 5.97 (s, 2H), 2.57 (s, 3H). LC/MS (ESI): 347.1 [M+H]+.

ACS Paragon Plus Environment

Page 50 of 91

Page 51 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

5-(1H-Indol-2-yl)-8-(methoxymethoxy)quinoline (5i) Whiter solid. 91% yield. Rf = 0.26 (petroleum/EtOAc = 1/1). 1H NMR (400 MHz, CDCl3) δ 8.97 (dd, J = 4.1, 1.7 Hz, 1H), 8.63 (dd, J = 8.6, 1.7 Hz, 1H), 8.47 (s, 1H), 7.69 (d, J = 7.8 Hz, 1H), 7.60 (d, J = 8.1 Hz, 1H), 7.48 – 7.39 (m, 3H), 7.27 – 7.22 (m, 1H), 7.18 (td, J = 7.6, 1.0 Hz, 1H), 6.71 (dd, J = 2.0, 0.8 Hz, 1H), 5.53 (s, 2H), 3.59 (s, 3H). LC/MS (ESI): 305.1 [M+H]+. 8-(Methoxymethoxy)-5-(5-(methoxymethoxy)-1H-indol-2-yl)quinoline (5j) Whiter solid. 93% yield. Rf = 0.22 (petroleum/EtOAc = 1/1). 1H NMR (400 MHz, CDCl3) δ 8.99 (dd, J = 4.1, 1.7 Hz, 1H), 8.64 (dd, J = 8.6, 1.7 Hz, 1H), 8.30 (s, 1H), 7.60 (d, J = 8.1 Hz, 1H), 7.47 (d, J = 7.9 Hz, 1H), 7.45 (dd, J = 8.4, 3.9 Hz, 1H), 7.36 (t, J = 5.6 Hz, 2H), 7.00 (dd, J = 8.7, 2.4 Hz, 1H), 6.64 (d, J = 1.3 Hz, 1H), 5.55 (s, 2H), 5.24 (s, 2H), 3.60 (s, 3H), 3.55 (s, 3H). LC/MS (ESI): 365.1 [M+H]+. 5,7-Dichloro-2-(1H-indol-2-yl)quinolin-8-yl acetate (6a) Yellow solid. 88% yield. Rf = 0.52 (petroleum/EtOAc = 10/1). 1H NMR (400 MHz, CDCl3) δ 9.36 (s, 1H), 8.46 (d, J = 8.9 Hz, 1H), 7.95 (d, J = 8.9 Hz, 1H), 7.68 (d, J = 7.9 Hz, 1H), 7.56 (s, 1H), 7.43 (d, J = 8.2 Hz, 1H), 7.29 (d, J = 7.1 Hz, 1H), 7.20 (d, J = 1.2 Hz, 1H), 7.17 – 7.08 (m, 1H), 2.64 (s, 3H). LC/MS (ESI): 371.0 [M+H]+. 5,7-Dichloro-2-(5-fluoro-1H-indol-2-yl)quinolin-8-yl acetate (6b) Yellow solid. 67% yield. Rf = 0.22 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 9.33 (s, 1H), 8.48 (d, J = 8.9 Hz, 1H), 7.93 (d, J = 8.9 Hz, 1H), 7.58 (s, 1H), 7.35 (dd, J = 9.0, 4.4 Hz, 1H), 7.31 (dd, J = 9.3, 2.4 Hz, 1H), 7.14 (d, J = 1.4 Hz, 1H), 7.04 (td, J = 9.0, 2.5 Hz, 1H), 2.63 (s, 3H). LC/MS (ESI): 389.0 [M+H]+.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

5,7-Dichloro-2-(5-chloro-1H-indol-2-yl)quinolin-8-yl acetate (6c) Yellow solid. 80% yield. Rf = 0.26 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 9.38 (s, 1H), 8.46 (d, J = 8.9 Hz, 1H), 7.91 (d, J = 8.8 Hz, 1H), 7.64 (s, 1H), 7.54 (s, 1H), 7.34 (d, J = 8.7 Hz, 1H), 7.22 (dd, J = 8.7, 1.3 Hz, 1H), 7.09 (s, 1H), 2.64 (s, 3H). LC/MS (ESI): 405.0 [M+H]+. 2-(5-Acetoxy-1H-indol-2-yl)-5,7-dichloroquinolin-8-ylacetate (6d) Yellow solid. 87% yield. Rf = 0.34 (petroleum/EtOAc = 2/1). 1H NMR (400 MHz, CDCl3) δ 9.33 (s, 1H), 8.43 (d, J = 8.9 Hz, 1H), 7.87 (d, J = 8.9 Hz, 1H), 7.51 (s, 1H), 7.36 (dd, J = 5.5, 3.2 Hz, 2H), 7.10 (d, J = 1.5 Hz, 1H), 6.99 (dd, J = 8.8, 2.1 Hz, 1H), 2.64 (s, 3H), 2.35 (s, 3H). LC/MS (ESI): 429.0 [M+H]+. 5,7-Dichloro-2-(5,6-dimethoxy-1H-indol-2-yl)quinolin-8-yl acetate (6e) Yellow solid. 82% yield. Rf = 0.27 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 9.20 (s, 1H), 8.31 (d, J = 8.9 Hz, 1H), 7.76 (d, J = 8.9 Hz, 1H), 7.40 (s, 1H), 7.05 (s, 1H), 6.97 (d, J = 1.5 Hz, 1H), 6.82 (s, 1H), 3.99 (s, 3H), 3.98 (s, 3H), 2.68 (s, 3H). LC/MS (ESI): 431.0 [M+H]+. 2-(8-Acetoxy-5,7-dichloroquinolin-2-yl)-6-methoxy-1H-indol-5-yl acetate (6f) Yellow solid. 85% yield. Rf = 0.26 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, DMSO) δ 8.54 (d, J = 9.0 Hz, 1H), 8.29 (d, J = 9.0 Hz, 1H), 7.99 (s, 1H), 7.42 (s, 1H), 7.35 (s, 1H), 7.18 (s, 1H), 3.84 (s, 3H), 2.67 (s, 3H), 2.28 (s, 3H). LC/MS (ESI): 459.0 [M+H]+. 2-(5H-[1,3]Dioxolo[4,5-f]indol-6-yl)-5,7-dichloroquinolin-8-yl acetate (6g)

ACS Paragon Plus Environment

Page 52 of 91

Page 53 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Yellow solid. 71% yield. Rf = 0.24 (petroleum/EtOAc = 5/1). 1H NMR (400 MHz, CDCl3) δ 9.24 (s, 1H), 8.41 (d, J = 9.0 Hz, 1H), 7.86 (d, J = 9.0 Hz, 1H), 7.56 (s, 1H), 7.07 (s, 1H), 7.01 (s, 1H), 6.86 (s, 1H), 5.99 (s, 2H), 2.62 (s, 3H). LC/MS (ESI): 415.0 [M+H]+. 7-Chloro-8-(methoxymethoxy)-5-(5-(methoxymethoxy)-1H-indol-2-yl)quinoline (6j) Whiter solid. 92% yield. Rf = 0.37 (petroleum/EtOAc = 3/1). 1H NMR (400 MHz, CDCl3) δ 8.91 (dd, J = 4.1, 1.7 Hz, 1H), 8.59 (dd, J = 8.6, 1.7 Hz, 1H), 8.39 (s, 1H), 7.66 (s, 1H), 7.40 (dd, J = 8.6, 4.1 Hz, 1H), 7.37 (d, J = 3.0 Hz, 1H), 7.36 (d, J = 3.4 Hz, 1H), 7.02 (dd, J = 8.8, 2.3 Hz, 1H), 6.66 (d, J = 1.3 Hz, 1H), 5.68 (s, 2H), 5.24 (s, 2H), 3.75 (s, 3H), 3.55 (s, 3H). LC/MS (ESI): 399.1 [M+H]+. General Procedures for the Synthesis of 7a-7g Compound 5a-5g (1.0 g, 1.0 equiv) and K2CO3 (1.0 equiv) in 15 ml CH3OH was stirred at room temperature. After the completion of the reaction, 100 mL of water was added; filtrated, washed with pure water, and dried under vacuum to get the crude product, which was purified by flash chromatography on silica gel and further recrystallized from CH3OH. 2-(1H-Indol-2-yl)quinolin-8-ol (7a) Yellow solid. 83% yield. Rf = 0.35 (petroleum/EtOAc = 1/1), mp 241.5-242.3 °C. 1H NMR (400 MHz, CDCl3) δ 9.54 (s, 1H), 8.13 (d, J = 8.7 Hz, 1H), 7.91 (d, J = 8.6 Hz, 1H), 7.69 (d, J = 8.0 Hz, 1H), 7.47 (dd, J = 8.2, 0.8 Hz, 1H), 7.43 – 7.36 (m, 1H), 7.34 – 7.25 (m, 2H), 7.22 – 7.16 (m, 2H), 7.16 – 7.09 (m, 1H). 13C NMR (101 MHz, CDCl3)

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

δ 151.76, 148.17, 137.48, 137.06, 136.73, 136.23, 128.99, 127.56, 127.28, 124.04, 121.54, 120.48, 119.16, 118.12, 111.43, 110.92, 103.18. FT-IR 3397, 3369, 3046, 1598, 1566, 1505, 1441, 1413, 1338, 1308, 1244, 1142, 1084, 831, 784, 753, 720, 661, 471; HRMS (ESI) m/z [M-H]- for C17H12N2O pred. 259.0877, meas. 259.0866; HPLC purity: 98.43%, retention time: 8.960 min. 2-(5-Fluoro-1H-indol-2-yl)quinolin-8-ol (7b) Yellow solid. 91% yield. Rf = 0.39 (petroleum/EtOAc = 1/1), mp 257.2-257.8 °C. 1H NMR (400 MHz, DMSO) δ 8.36 (d, J = 8.6 Hz, 1H), 8.13 (d, J = 8.6 Hz, 1H), 7.53 (dd, J = 8.7, 4.4 Hz, 1H), 7.49 – 7.38 (m, 3H), 7.36 (s, 1H), 7.16 (d, J = 7.1 Hz, 1H), 7.10 (td, J = 9.3, 2.5 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 158.80 (s), 156.49 (s), 153.47 (s), 147.83 (s), 139.31 (s), 137.76 (s), 137.32 (s), 134.24 (s), 129.19 (d, J = 10.5 Hz), 127.95 (s), 119.21 (s), 117.95 (s), 112.99 (d, J = 9.7 Hz), 112.04 (d, J = 26.3 Hz), 111.52 (s), 105.97 (d, J = 23.1 Hz), 102.56 (d, J = 4.8 Hz). FT-IR 3390, 3364, 3051, 1628, 1599, 1567, 1549, 1507, 1494, 1442, 1418, 1315, 1286, 1243, 1201, 1129, 1110, 953, 832, 782, 752, 717, 657, 605, 566, 484, 462; HRMS (ESI) m/z [M+H]+ for C17H11FN2O pred. 279.0928, meas. 279.0929; HPLC purity: 95.24%, retention time: 7.403 min. 2-(5-Chloro-1H-indol-2-yl)quinolin-8-ol (7c) Yellow solid. 86% yield. Rf = 0.41 (petroleum/EtOAc = 1/1), mp 305.5-306.3 °C. 1H NMR (400 MHz, DMSO) δ 8.37 (d, J = 8.6 Hz, 1H), 8.14 (d, J = 8.6 Hz, 1H), 7.70 (s, 1H), 7.53 (d, J = 8.6 Hz, 1H), 7.48 – 7.39 (m, 2H), 7.36 (s, 1H), 7.23 (d, J = 8.6 Hz, 1H), 7.15 (d, J = 7.1 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 153.40, 147.70, 139.06,

ACS Paragon Plus Environment

Page 54 of 91

Page 55 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

137.71, 137.35, 135.90, 130.13, 128.01, 127.97, 124.61, 123.64, 120.66, 119.23, 118.02, 113.48, 111.52, 102.09. FT-IR 3374, 3328, 2923, 1599, 1569, 1547, 1506, 1440, 1471, 1412, 1318, 1245, 1225, 1183, 1142, 1083, 1058, 915, 832, 778, 748, 716, 689, 655, 605, 571, 533, 476; HRMS (ESI) m/z [M+H]+ for C17H11ClN2O pred. 295.0633, meas. 295.0630; HPLC purity: 97.89%, retention time: 7.495 min. 2-(5-Hydroxy-1H-indol-2-yl)quinolin-8-ol (7d) Yellow solid. 87% yield. Rf = 0.31 (petroleum/EtOAc = 1/1), mp 282.7-283.1 °C. 1H NMR (400 MHz, DMSO) δ 8.31 (d, J = 8.7 Hz, 1H), 8.07 (d, J = 8.7 Hz, 1H), 7.43 – 7.35 (m, 2H), 7.30 (d, J = 8.7 Hz, 1H), 7.17 (s, 1H), 7.11 (dd, J = 7.0, 1.6 Hz, 1H), 6.94 (d, J = 2.0 Hz, 1H), 6.77 (dd, J = 8.7, 2.2 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 152.61 (s), 150.84 (s), 147.82 (s), 137.18 (d, J = 6.1 Hz), 137.07 (s), 136.49 (s), 131.75 (d, J = 14.8 Hz), 129.21 (d, J = 4.4 Hz), 127.13 (s), 126.95 (s), 118.58 (s), 117.43 (s), 113.97 (d, J = 9.7 Hz), 111.76 (d, J = 5.1 Hz), 110.75 (d, J = 5.3 Hz), 104.33 (d, J = 7.8 Hz), 101.29 (d, J = 4.6 Hz). FT-IR 3480, 3399, 3357, 1708, 1632, 1598, 1566, 1548, 1504, 1442, 1420, 1338, 1314, 1242, 1210, 1135, 1081, 949, 834, 783, 753, 719, 665, 618, 467; HRMS (ESI) m/z [M-H]- for C17H12N2O2 pred. 275.0826, meas. 275.0820; HPLC purity: 97.30%, retention time: 4.914 min. 2-(5,6-Dimethoxy-1H-indol-2-yl)quinolin-8-ol (7e) Yellow solid. 83% yield. Rf = 0.32 (petroleum/EtOAc = 1/1), mp 320.1-320.7 °C.1H NMR (400 MHz, DMSO) δ 11.89 (s, 1H), 8.26 (d, J = 8.7 Hz, 1H), 8.00 (d, J = 8.7 Hz, 1H), 7.47 – 7.32 (m, 2H), 7.21 – 7.13 (m, 2H), 7.11 (s, 1H), 7.00 (s, 1H), 3.88 (s, 3H), 3.81 (s, 3H). 13C NMR (101 MHz, DMSO) δ 153.17, 148.70, 148.45, 145.54,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

137.79, 136.90, 135.99, 132.57, 127.44, 127.20, 121.87, 118.87, 117.94, 111.31, 103.21, 102.78, 94.78, 56.20, 55.98. FT-IR 3426, 3381, 3005, 2933, 1626, 1598, 1566, 1544, 1509, 1483, 1452, 1440, 1406, 1390, 1347, 1317, 1287, 1245, 1211, 1133, 1006, 887, 849, 833, 796, 743, 720, 651, 600, 572, 476, 457; HRMS (ESI) m/z [M+H]+ for C19H16N2O3 pred. 321.1234, meas. 321.1224; HPLC purity: 99.23%, retention time: 7.600 min. 2-(5-Hydroxy-6-methoxy-1H-indol-2-yl)quinolin-8-ol (7f) Yellow solid. 81% yield. Rf = 0.28 (petroleum/EtOAc = 1/1), mp 302.2-302.9 °C. 1H NMR (400 MHz, DMSO) δ 11.75 (s, 1H), 9.64 (s, 1H), 8.10 (d, J = 8.7 Hz, 1H), 7.83 (d, J = 8.7 Hz, 1H), 7.70 (d, J = 3.0 Hz, 1H), 7.36 – 7.31 (m, 1H), 7.28 (d, J = 7.8 Hz, 1H), 7.07 (d, J = 7.2 Hz, 1H), 7.03 (s, 1H), 6.63 (s, 1H), 3.99 (s, 3H). 13C NMR (101 MHz, DMSO) δ 153.12, 148.66, 148.18, 139.59, 137.82, 136.71, 135.33, 131.78, 127.33, 126.94, 123.24, 118.95, 117.89, 115.15, 111.15, 103.22, 93.52, 56.38. FT-IR 3476, 3413, 3353, 3018, 2947, 1662, 1587, 1558, 1531, 1511, 1479, 1447, 1436, 1418, 1386, 1351, 1309, 1290, 1268, 1209, 1142, 1062, 879, 852, 841, 789, 745, 728, 649, 606, 574, 469, 458; HRMS (ESI) m/z [M+H]+ for C18H12N2O3 pred. 307.1077, meas. 307.1063; HPLC purity: 95.80%, retention time: 7.385 min. 2-(8H-[1,3]Dioxolo[4,5-g]indol-7-yl)quinolin-8-ol (7g) Yellow solid. 76% yield. Rf = 0.36 (petroleum/EtOAc = 1/1), mp 273.2-273.9 °C. 1H NMR (400 MHz, DMSO) δ 8.28 (d, J = 8.7 Hz, 1H), 8.01 (d, J = 8.6 Hz, 1H), 7.37 (q, J = 8.3 Hz, 2H), 7.22 (s, 1H), 7.10 (d, J = 6.6 Hz, 2H), 6.99 (s, 1H), 6.00 (s, 2H). 13C NMR (101 MHz, DMSO) δ 152.61 (d, J = 14.3 Hz), 147.78 (s), 145.76 (s), 142.78 (s),

ACS Paragon Plus Environment

Page 56 of 91

Page 57 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

137.25 (d, J = 6.3 Hz), 136.46 (s), 135.73 (d, J = 13.7 Hz), 132.54 (d, J = 14.9 Hz), 126.96 (s), 126.72 (s), 122.50 (d, J = 4.2 Hz), 118.30 (s), 117.46 (d, J = 2.3 Hz), 110.77 (d, J = 5.6 Hz), 102.74 (d, J = 3.5 Hz), 100.44 (s), 99.12 (s), 91.81 (d, J = 5.2 Hz). FT-IR 3357, 2893, 1598, 1564, 1543, 1504, 1468, 1416, 1350, 1315, 1284, 1239, 1211, 1086, 1036, 948, 829, 782, 751, 717, 663, 451; HRMS (ESI) m/z [M+H]+ for C18H14N2O3 pred. 305.0921, meas. 305.0911; HPLC purity: 98.66%, retention time: 10.282 min. General Procedures for the Synthesis of 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl. Compound 7g, 8c, 8d or 8g (1.0 g, HPLC purity > 95%) was dissolved in 30 mL of EtOAc, and then HCl gas was bubbled into the solution at room temperature for 12 h. The mixture was filtrated and the precipitation was washed with 40 mL of EtOAc, dried under reduced pressure to obtain an orange solid which was used in the vivo assay, pharmacokinetics and metabolic stability study. 2-(8H-[1,3]Dioxolo[4,5-g]indol-7-yl)quinolin-8-ol hydrochloride (7g•HCl) Yellow solid. 93% yield. Rf = 0.36 (petroleum/EtOAc = 1/1), mp 299.2-299.9 °C. 1H NMR (400 MHz, DMSO) δ 12.11 (s, 1H), 8.34 (d, J = 8.7 Hz, 1H), 8.06 (d, J = 8.7 Hz, 1H), 7.44 – 7.34 (m, 2H), 7.29 (d, J = 1.8 Hz, 1H), 7.15 (dd, J = 7.0, 1.8 Hz, 1H), 7.09 (s, 1H), 7.00 (s, 1H), 6.01 (s, 2H).

13

C NMR (101 MHz, DMSO) δ 152.29 (s),

147.72 (s), 146.80 (s), 143.54 (s), 138.17 (s), 136.10 (d, J = 5.1 Hz), 134.72 (d, J = 7.8 Hz), 133.72 (d, J = 17.8 Hz), 127.52 (d, J = 10.3 Hz), 122.88 (dd, J = 8.7, 4.0 Hz), 119.15 (s), 118.13 (s), 112.28 (s), 105.00 (d, J = 7.0 Hz), 101.10 (s), 99.57 (d, J = 2.5 Hz), 92.37 (d, J = 5.5 Hz). FT-IR 3356, 2892, 1599, 1568, 1547, 1504, 1469, 1416,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1355, 1316, 1285, 1241, 1213, 1087, 1036, 949, 829, 783, 750, 718, 662, 453; HRMS (ESI) m/z [M+H]+ for C18H14N2O3 pred. 305.0921, meas. 305.0904; HPLC purity: 99.88%, retention time: 15.780 min. General Procedures for the Synthesis of 8a-8g Compound 6a-6g (1.0 g, 1.0 equiv) and NaOCH3 (6.0 equiv) in 15 mL of CH3OH was stirred at room temperature. After the completion of the reaction, 100 mL of water was added, and then the mixture was filtrated, washed with pure water, and dried under vacuum to get the crude product, which was purified by flash chromatography on silica gel and further recrystallized from CH3OH. 5,7-Dichloro-2-(1H-indol-2-yl)quinolin-8-ol (8a) Yellow solid. 93% yield. Rf = 0.36 (petroleum/EtOAc = 1/1), mp 265.0-265.9 °C. 1H NMR (400 MHz, DMSO) δ 12.10 (s, 1H), 8.36 (d, J = 8.7 Hz, 1H), 8.18 (d, J = 8.7 Hz, 1H), 7.67 (d, J = 7.1 Hz, 2H), 7.54 (d, J = 8.1 Hz, 1H), 7.38 (s, 1H), 7.29 (t, J = 7.5 Hz, 1H), 7.11 (t, J = 7.4 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 149.70 (d, J = 2.6 Hz), 149.10 (s), 138.43 (s), 137.71 (d, J = 15.6 Hz), 136.53 (d, J = 13.8 Hz), 133.72 (s), 128.89 (d, J = 4.4 Hz), 127.39 (s), 124.32 (s), 123.75 (s), 121.95 (s), 120.32 (s), 119.62 (s), 115.58 (s), 112.05 (d, J = 5.6 Hz), 104.00 (d, J = 3.5 Hz). FT-IR 3524, 3401, 3319, 3059, 1657, 1617, 1596, 1545, 1493, 1435, 1415, 1392, 1340, 1306, 1250, 1218, 1182, 1146, 982, 948, 890, 858, 829, 785, 746, 725, 625, 560, 487, 441; HRMS (ESI) m/z [M+H]+ for C17H10Cl2N2O pred. 329.0242, meas. 329.0243; HPLC purity: 98.28%, retention time: 7.370 min. 5,7-Dichloro-2-(5-fluoro-1H-indol-2-yl)quinolin-8-ol (8b)

ACS Paragon Plus Environment

Page 58 of 91

Page 59 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Yellow solid. 84% yield. Rf = 0.31 (petroleum/EtOAc = 1/1), mp 278.7-279.4 °C. 1H NMR (400 MHz, DMSO) δ 8.48 (d, J = 8.9 Hz, 1H), 8.29 (d, J = 8.9 Hz, 1H), 7.76 (s, 1H), 7.52 (dd, J = 8.9, 4.5 Hz, 1H), 7.45 – 7.42 (m, 2H), 7.11 (td, J = 9.2, 2.5 Hz, 1H). 13

C NMR (101 MHz, DMSO) δ 157.68 (d, J = 232.9 Hz), 149.47 (s), 149.15 (s),

138.43 (s), 138.27 (d, J = 14.5 Hz), 134.45 (d, J = 15.6 Hz), 134.00 (s), 129.00 (dd, J = 10.6, 3.8 Hz), 127.65 (s), 123.92 (s), 120.43 (s), 119.67 (s), 115.66 (s), 113.21 (dd, J = 9.1, 5.5 Hz), 112.70 (d, J = 26.4 Hz), 106.15 (d, J = 23.3 Hz), 103.95 (d, J = 3.9 Hz). FT-IR 3521, 3292, 3069, 1614, 1595, 1545, 1490, 1435, 1417, 1343, 1306, 1254, 1223, 1199, 1146, 1111, 985, 952, 850, 829, 780, 722, 826, 594, 557, 496, 484, 440; HRMS (ESI) m/z [M+H]+ for C17H9Cl2FN2O pred. 347.0149, meas. 347.0152; HPLC purity: 96.21%, retention time: 7.366 min. 5,7-Dichloro-2-(5-chloro-1H-indol-2-yl)quinolin-8-ol (8c) Yellow solid. 77% yield. Rf = 0.37 (petroleum/EtOAc = 1/1), mp 293.1-294.0 °C. 1H NMR (400 MHz, DMSO) δ 12.26 (s, 1H), 8.47 (d, J = 0.9 Hz, 1H), 8.28 (d, J = 2.7 Hz, 1H), 7.73 (d, J = 13.7 Hz, 2H), 7.53 (d, J = 7.6 Hz, 1H), 7.43 (s, 1H), 7.24 (d, J = 7.9 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 163.11, 150.21, 146.34, 144.68, 143.43, 132.61, 131.94, 127.65, 124.99, 121.34, 119.50, 119.36, 118.70, 116.98, 115.73, 105.10, 99.85. FT-IR 3594, 3516, 3438, 3318, 3074, 1717, 1618, 1595, 1559, 1542, 1493, 1471, 1435, 1410, 1390, 1341, 1305, 1255, 1218, 1178, 1146, 1060, 983, 948, 918, 864, 851, 831, 792, 746, 721, 690, 629, 557, 490, 440; HRMS (ESI) m/z [M+H]+ for C17H9Cl3N2O pred. 362.9853, meas. 362.9839; HPLC purity: 99.37%, retention time: 9.760 min.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

5,7-Dichloro-2-(5-chloro-1H-indol-2-yl)quinolin-8-ol hydrochloride (8c•HCl) Yellow solid. 90% yield. Rf = 0.37 (petroleum/EtOAc = 1/1), mp 304.1-304.9 °C. 1H NMR (400 MHz, DMSO) δ 8.08 (d, J = 8.7 Hz, 1H), 7.90 (d, J = 8.8 Hz, 1H), 7.33 (s, 1H), 7.32 – 7.19 (m, 2H), 6.91 (s, 1H), 6.67 (d, J = 8.4 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 164.02, 151.05, 147.26, 146.50, 143.25, 136.24, 131.01, 129.57, 125.01, 120.43, 119.89, 119.62, 118.09, 117.06, 116.07, 105.01, 100.29. FT-IR 3593, 3512, 3439, 3317, 3073, 1718, 1617, 1596, 1559, 1542, 1493, 1472, 1434, 1412, 1391, 1342, 1306, 1257, 1219, 1178, 1145, 1061, 984, 949, 918, 865, 852, 832, 791, 747, 722, 691, 629, 558, 491, 443; HRMS (ESI) m/z [M+H]+ for C17H9Cl3N2O pred. 362.9853, meas. 362.9836; HPLC purity: 99.91%, retention time: 15.996 min. 5,7-Dichloro-2-(5-hydroxy-1H-indol-2-yl)quinolin-8-ol (8d) Yellow solid. 91% yield. Rf = 0.36 (petroleum/EtOAc = 1/1), mp 276.4-277.6 °C. 1H NMR (400 MHz, DMSO) δ 8.31 (d, J = 8.7 Hz, 8H), 8.07 (d, J = 8.7 Hz, 8H), 7.43 – 7.35 (m, 16H), 7.30 (d, J = 8.7 Hz, 8H), 7.17 (s, 8H), 7.11 (dd, J = 7.0, 1.6 Hz, 8H), 6.94 (d, J = 2.0 Hz, 8H), 6.77 (dd, J = 8.7, 2.2 Hz, 8H). 13C NMR (101 MHz, DMSO) δ 152.61 (s), 150.84 (s), 147.82 (s), 137.18 (d, J = 6.1 Hz), 137.07 (s), 136.49 (s), 131.75 (d, J = 14.8 Hz), 129.21 (d, J = 4.4 Hz), 127.13 (s), 126.95 (s), 118.58 (s), 117.43 (s), 113.97 (d, J = 9.7 Hz), 111.76 (d, J = 5.1 Hz), 110.75 (d, J = 5.3 Hz), 104.33 (d, J = 7.8 Hz), 101.29 (d, J = 4.6 Hz). FT-IR 3547, 3464, 3302, 3213, 2777, 1711, 1594, 1542, 1493, 1434, 1396, 1305, 1260, 1226, 1199, 1147, 1083, 983, 951, 854, 835, 783, 721, 628, 493, 426; HRMS (ESI) m/z [M-H]- for C17H10N2O2Cl2 pred. 343.0047, meas. 343.0034; HPLC purity: 99.35%, retention time: 8.807 min.

ACS Paragon Plus Environment

Page 60 of 91

Page 61 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

5,7-Dichloro-2-(5-hydroxy-1H-indol-2-yl)quinolin-8-ol hydrochloride (8d•HCl) Yellow solid. 96% yield. Rf = 0.36 (petroleum/EtOAc = 1/1), mp 312.7-313.3 °C. 1H NMR (400 MHz, DMSO) δ 8.43 (d, J = 8.9 Hz, 1H), 8.22 (d, J = 8.9 Hz, 1H), 7.72 (s, 1H), 7.34 (d, J = 8.7 Hz, 1H), 7.27 (s, 1H), 6.97 (d, J = 2.1 Hz, 1H), 6.82 (dd, J = 8.7, 2.2 Hz, 1H). 13C NMR (101 MHz, DMSO) δ 151.61 (s), 150.00 (d, J = 3.6 Hz), 148.99 (s), 138.49 (s), 136.68 (dd, J = 14.2, 7.2 Hz), 133.71 (s), 132.61 (d, J = 15.5 Hz), 129.58 (dd, J = 8.7, 4.3 Hz), 127.24 (s), 123.67 (s), 120.40 (s), 119.70 (s), 115.52 (s), 115.29 (s), 112.58 (d, J = 5.6 Hz), 104.92 (s), 103.24 (d, J = 3.8 Hz). FT-IR 3546, 3466, 3306, 3218, 2776, 1714, 1597, 1543, 1496, 1438, 1397, 1305, 1260, 1228, 1199, 1149, 1085, 983, 958, 857, 836, 787, 726, 628, 495, 428; HRMS (ESI) m/z [M+H]+ for C17H10Cl2N2O2 pred. 345.0192, meas. 345.0179; HPLC purity: 99.58%, retention time: 26.961min. 5,7-Dichloro-2-(5,6-dimethoxy-1H-indol-2-yl)quinolin-8-ol (8e) Yellow solid. 75% yield. Rf = 0.34 (petroleum/EtOAc = 1/1), mp 245.8-246.4 °C. 1H NMR (400 MHz, DMSO) δ 12.03 (s, 1H), 8.33 (d, J = 8.6 Hz, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.65 (s, 1H), 7.22 (s, 1H), 7.09 (s, 1H), 6.94 (s, 1H), 3.85 (s, 3H), 3.80 (s, 3H). 13C NMR (101 MHz, DMSO) δ 149.77, 149.09, 145.67, 138.91, 133.42, 132.96, 132.80, 127.04, 126.90, 123.49, 121.77, 120.00, 115.50, 104.07, 103.85, 103.12, 94.72, 56.18, 55.95. FT-IR 3583, 3524, 3313, 3246, 3075, 2998, 2934, 2834, 1595, 1541, 1484, 1462, 1436, 1412, 1350, 1294, 1250, 1210, 1141, 1082, 1008, 984, 947, 852, 812, 751, 714, 622, 516, 485, 438; HRMS (ESI) m/z [M+H]+ for C19H14Cl2N2O3 pred. 389.0454, meas. 389.0450; HPLC purity: 99.45%, retention time: 7.580 min.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

5,7-Dichloro-2-(5-hydroxy-6-methoxy-1H-indol-2-yl)quinolin-8-ol (8f) Yellow solid. 69% yield. Rf = 0.26 (petroleum/EtOAc = 1/1), mp 310.9-311.3 °C. 1H NMR (400 MHz, DMSO) δ 11.78 (s, 1H), 10.42 (s, 1H), 8.22 (d, J = 9.0 Hz, 1H), 8.01 (d, J = 9.0 Hz, 1H), 7.78 (s, 1H), 7.64 (s, 1H), 7.04 (s, 1H), 6.73 (d, J = 0.9 Hz, 1H), 4.00 (s, 3H). 13C NMR (101 MHz, DMSO) δ 150.21 (d, J = 1.6 Hz), 148.80 (d, J = 2.6 Hz), 148.69 (d, J = 4.1 Hz), 139.82 (d, J = 12.8 Hz), 138.60 (d, J = 7.6 Hz), 134.40 (s), 133.30 (s), 132.23 (s), 126.69 (s), 123.30 (s), 123.21 (s), 120.34 (s), 119.74 (d, J = 5.7 Hz), 115.36 (d, J = 3.0 Hz), 114.96 (d, J = 6.8 Hz), 104.81 (s), 93.45 (s), 56.37 (s). FT-IR 3589, 3546, 3332, 3265, 3056, 2989, 2941, 2840, 1558, 1516, 1449, 1426, 1406, 1392, 1346, 1289, 1248, 1209, 1135, 1078, 1003, 979, 951, 869, 822, 753, 712, 626, 511, 483, 434; HRMS (ESI) m/z [M+H]+ for C18H11Cl2N2O3 pred. 375.0298 meas. 375.0292; HPLC purity: 95.18%, retention time: 5.112 min. 2-(5H-[1,3]Dioxolo[4,5-f]indol-6-yl)-5,7-dichloroquinolin-8-ol (8g) Yellow solid. 79% yield. Rf = 0.35 (petroleum/EtOAc = 1/1), mp 292.9-293.3 °C. 1H NMR (400 MHz, DMSO) δ 8.29 (d, J = 8.7 Hz, 1H), 8.03 (d, J = 8.7 Hz, 1H), 7.58 (s, 1H), 7.17 (s, 1H), 7.04 (s, 1H), 6.91 (s, 1H), 5.98 (s, 2H). 13C NMR (101 MHz, DMSO) δ 148.99, 146.36, 143.31, 140.10, 135.97, 135.82, 133.42, 133.34, 133.27, 127.22, 123.89, 122.92, 119.67, 115.71, 103.99, 100.97, 99.52, 92.46. FT-IR 3307, 3111, 3074, 2889, 1595, 1541, 1499, 1471, 1440, 1416, 1349, 1309, 1284, 1242, 1210, 1185, 1151, 1040, 983, 947, 844, 826, 783, 745, 722, 683, 624, 560, 501, 463; HRMS (ESI) m/z [M+H]+ for C18H10Cl2N2O3 pred. 373.0141, meas. 373.0137; HPLC purity: 99.22%, retention time: 12.052 min.

ACS Paragon Plus Environment

Page 62 of 91

Page 63 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

2-(5H-[1,3]Dioxolo[4,5-f]indol-6-yl)-5,7-dichloroquinolin-8-ol

hydrochloride

(8g•HCl) Yellow solid. 92% yield. Rf = 0.35 (petroleum/EtOAc = 1/1), mp 321.9-322.3 °C. 1H NMR (400 MHz, DMSO) δ 8.39 (d, J = 8.9 Hz, 1H), 8.17 (d, J = 8.9 Hz, 1H), 7.69 (s, 1H), 7.31 (s, 1H), 7.10 (s, 1H), 7.01 (s, 1H), 6.02 (s, 2H). 13C NMR (101 MHz, DMSO) δ 149.79 (d, J = 3.3 Hz), 148.88 (s), 146.72 (s), 143.46 (s), 138.51 (s), 135.22 (dd, J = 14.5, 7.2 Hz), 133.49 (dd, J = 11.2, 4.5 Hz), 126.87 (s), 123.39 (s), 122.84 (dd, J = 8.5, 4.0 Hz), 120.04 (s), 119.68 (s), 115.45 (s), 104.64 (d, J = 3.1 Hz), 101.07 (s), 99.63 (s), 92.34 (d, J = 5.6 Hz). FT-IR 3316, 3112, 3078, 2891, 1596, 1543, 1499, 1473, 1442, 1417, 1349, 1309, 1286, 1245, 1213, 1186, 1154, 1041, 984, 949, 846, 828, 786, 747, 726, 685, 628, 561, 503, 467; HRMS (ESI) m/z [M+H]+ for C18H10Cl2N2O3 pred. 373.0141, meas. 373.0132; HPLC purity: 99.96%, retention time: 17.382 min. General Procedures for the Synthesis of 9a, 9d and 10d

To a solution of 9a, 9d or 10d (1.0 g, 1.0 equiv) in 10 mL of CH3OH, 2 mL 37% hydrochloric acid was added. After stirred at room temperature for 2 h, the reaction mixture was filtrated and the residue was dissolved in 200 mL saturated NaHCO3 solution. Then, filtrated, washed with pure water and dried under vacuum to get the crude product which was further purified by recrystallization from AcOEt.

5-(1H-Indol-2-yl)quinolin-8-ol (9a)

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Yellow solid. 81% yield. Rf = 0.26 (CH2Cl2/CH3OH = 10/1), mp 194.8-195.1 °C. 1H NMR (400 MHz, CDCl3) δ 8.83 (dd, J = 4.1, 1.5 Hz, 1H), 8.68 (dd, J = 8.6, 1.5 Hz, 1H), 8.26 (s, 1H), 7.69 (d, J = 7.8 Hz, 1H), 7.63 (d, J = 7.9 Hz, 1H), 7.49 (dd, J = 8.6, 4.2 Hz, 1H), 7.45 (d, J = 8.1 Hz, 1H), 7.26 – 7.22 (m, 2H), 7.22 – 7.14 (m, 1H), 6.70 (s, 1H). 13C NMR (101 MHz, CDCl3) δ 152.38, 148.03, 138.36, 136.40, 135.47, 134.59, 128.99, 128.32, 126.90, 122.31, 122.26, 121.84, 120.53, 120.32, 110.86, 109.56, 103.35. FT-IR 3342, 3054, 2506, 1880, 1580, 1500, 1469, 1454, 1413, 1366, 1343, 1297, 1271, 1226, 1190, 1158, 1094, 924, 903, 840, 790, 742, 694, 654, 642, 566, 517, 492, 435; HRMS (ESI) m/z [M-H]- for C17H12N2O pred. 259.0877, meas. 259.0868; HPLC purity: 95.83%, retention time: 5.862 min. 5-(5-Hydroxy-1H-indol-2-yl)quinolin-8-ol (9d) Yellow solid. 85% yield. Rf = 0.22 (CH2Cl2/CH3OH = 10/1), mp 235.7-236.3 °C. 1H NMR (400 MHz, DMSO) δ 8.91 (dd, J = 4.0, 1.4 Hz, 1H), 8.69 (dd, J = 8.6, 1.4 Hz, 1H), 7.73 – 7.57 (m, 2H), 7.21 (t, J = 8.6 Hz, 2H), 6.92 (d, J = 2.1 Hz, 1H), 6.66 (dd, J = 8.6, 2.2 Hz, 1H), 6.47 (s, 1H). 13C NMR (101 MHz, DMSO) δ 153.45 (s), 151.22 (d, J = 11.8 Hz), 148.67 (s), 138.91 (s), 136.56 (d, J = 14.6 Hz), 134.74 (s), 131.63 (d, J = 15.0 Hz), 129.71 (d, J = 3.5 Hz), 128.66 (s), 127.02 (s), 122.74 (s), 122.05 (d, J = 5.4 Hz), 112.00 (d, J = 4.5 Hz), 111.33 (s), 104.11 (d, J = 6.1 Hz), 101.60 (d, J = 4.2 Hz). FT-IR 3404, 3335, 2922, 1702, 1624, 1581, 1504, 1465, 1453, 1413, 1365, 1270, 1223, 1179, 1097, 1072, 953, 837, 785, 714, 647, 592, 566, 485, 459; HRMS (ESI) m/z [M+H]+ for C17H12N2O2 pred. 277.0972, meas. 277.0971; HPLC purity: 99.25%, retention time: 14.006 min.

ACS Paragon Plus Environment

Page 64 of 91

Page 65 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

7-Chloro-5-(5-hydroxy-1H-indol-2-yl)quinolin-8-ol (10d) Yellow solid. 90% yield. Rf = 0.29 (CH2Cl2/CH3OH = 20/1), mp 325.0-326.1 °C. 1H NMR (400 MHz, DMSO) δ 8.56 (d, J = 8.4 Hz, 2H), 7.55 (s, 1H), 7.34 (dd, J = 8.5, 4.1 Hz, 1H), 7.13 (d, J = 8.5 Hz, 1H), 6.83 (d, J = 2.1 Hz, 1H), 6.56 (dd, J = 8.5, 2.2 Hz, 1H), 6.28 (s, 1H). 13C NMR (101 MHz, DMSO) δ 163.64, 151.06, 146.03, 145.22, 138.25, 134.26, 131.29, 131.15, 130.08, 127.59, 121.12, 116.76, 111.55, 110.86, 108.57, 103.81, 99.63. FT-IR 3404, 3211, 1623, 1592, 1551, 1521, 1500, 1449, 1377, 1355, 1306, 1251, 1196, 1165, 1101, 871, 801, 785, 732, 684, 649, 611, 590, 501, 460; HRMS (ESI) m/z [M+H]+ for C17H11N2O2Cl pred. 311.0582, meas. 311.0594; HPLC purity: 95.22%, retention time: 9.986 min. Oxygen Radical Absorbance Capacity (ORAC-FL) Assay42, 43 The tested compound and fluorescein (FL) stock solution were diluted with 75 mM phosphate buffer (pH 7.4) to 10 µM (or 20 µM) and 0.117 µM, respectively. The solution of (±)-6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox) was diluted with the same buffer to 100, 80, 60, 50, 40, 20, and 10 µM. The solution of 2,2'-azobis-(amidinopropane)dihydrochloride (AAPH) was prepared by dissolving 108.4 mg AAPH in 10 mL 75 mM phosphate buffer (pH 7.4) to a final concentration of 40 mM. The mixture of the tested compound (20 µL) and FL (120 µL; 70 nM, final concentration) was pre-incubated for 10 min at 37ºC, and then 60 µL of the AAPH solution was added. The fluorescence was recorded every minute for 120 min (excitation, 485 nm; emission, 520 nm). A blank using phosphate buffer instead of the tested compound was also carried out. All reaction mixtures were prepared triple and

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

at least three independent runs were performed for each sample. The Antioxidant curves (fluorescence versus time) were normalized to the curve of the blank. The area under the fluorescence decay curve (AUC) was calculated as following equation: ௜ୀଵଶ଴

AUC = 1 + ෍ ሺfi/f0ሻ ௜ୀଵ

Where f0 is the initial fluorescence reading at 0 min and fi is the fluorescence reading at a specific time. The net AUC was calculated by the expression: AUCsample – AUCblank. Regression equations between net AUC and Trolox concentrations were calculated. ORAC-FL value for each sample were calculated by using the standard curve which means the ORAC-FL value of tested compound expressed as Trolox equivalents. Blood-Brain Barrier Permeation Assay The blood-brain barrier penetration of compounds was evaluated using the parallel artificial membrane permeation assay (PAMPA) described by Di et al.44-46 Commercial drugs were purchased from Sigma and Alfa Aesar. Porcine brain lipid (PBL) was obtained from Avanti Polar Lipids. The donor microplate (PVDF membrane, pore size 0.45 mm) and acceptor microplate were both from Millipore. The 96-well UV plate (COSTAR®) was from Corning Incorporated. The acceptor 96-well microplate was filled with 300 µL PBS/EtOH (7:3), and the filter membrane was impregnated with 4 µL PBL in dodecane (20 mg/mL). Compounds were dissolved in DMSO at 5 mg/mL and diluted 50-fold in PBS/EtOH (7:3) to a final concentration of 100 µg/mL. Then, 200 µL of the solution was added to the donor wells. The acceptor filter plate was carefully placed on the donor plate to form a

ACS Paragon Plus Environment

Page 66 of 91

Page 67 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

sandwich, which was left undisturbed for 10 h at 25 °C. After incubation, the donor plate was carefully removed, and the concentration of compounds in the acceptor wells was determined using the UV plate reader (Flexstation® 3). Every sample was analyzed at five wavelengths in four wells and in at least three independent runs. Pe was calculated by the following expression: Pe = -Vd×Va/[(Vd+Va)A×t] × ln(1-drugacceptor/drugequilibrium) where Vd is the volume of donor well; Va, volume in acceptor well; A, filter area; t, permeation time; drugacceptor, the absorbance obtained in the acceptor well; drugequilibrium, the theoretical equilibrium absorbance. The results are given as the mean ± standard deviation. In the experiment, 13 quality control standards (Table S1, see Supporting Information) of known BBB permeability were included to validate the analysis set. A plot of the experimental data versus literature values gave a strong linear correlation, Pe (exp.) = 1.4574Pe (lit.) - 1.0773 (R2 = 0.9427) (Figure S2). From this equation and the limit established by Di et al. ( Pe (lit.) = 4.0× 10-6 cm/s) for blood-brain barrier permeation, we concluded that compounds with a permeability greater than 4.7 × 10-6 cm/s could cross the blood-brain barrier (Table S2). ThT Assay47 Aβ1–42 (Millipore, counter ion: NaOH) was dissolved in ammonium hydroxide (1% v/v) to give a stock solution (2000 µM), which was aliquoted into small samples and stored at -80 °C. For the experiment of self-mediated Aβ1–42 aggregation inhibition, the Aβ stock solution was diluted with 50 mM phosphate buffer (pH 7.4) to 50 µM before use. A

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

mixture of the peptide (10 µL, 25 µM, final concentration) with or without the tested compound (10 µL) was incubated at 37 °C for 48 h. Blanks using 50 mM phosphate buffer (pH 7.4) instead of Aβ with or without inhibitors were also carried out. The sample was diluted to a final volume of 200 µL with 50 mM glycine–NaOH buffer (pH 8.0) containing thioflavin T (5 µM). Then the fluorescence intensities were recorded five minutes later (excitation, 450 nm; emission, 485 nm). The percent inhibition of aggregation was calculated by the expression (1-IFi/IFc) × 100%,in which IFi and IFc are the fluorescence intensities obtained for Aβ in the presence and absence of inhibitors after subtracting the background, respectively. For the experiment of copper-mediated Aβ1–42 aggregation inhibition, the Aβ stock solution was diluted with the solution of 20 µM HEPES (pH 6.6) and 150 µM NaCl. The mixture of the peptide (10 µL, 25 µM, final concentration) with or without copper (10 µL, 25 µM, final concentration) and the tested compound (10 µL, 50 µM, final concentration) was incubated at 37 °C for 24 h. Then 20 µL of the sample was diluted to a final volume of 200 µL with 50 mM glycine–NaOH buffer (pH 8.0) containing thioflavin T (5 µM). The detection method was the same as that of self-mediated Aβ1–42 aggregation experiment. For the experiment of self-induced Aβ fibrils disaggregation, the Aβ stock solution was diluted with 10 mM phosphate buffer (pH 7.4). The peptide (15µL, 50 µM) was incubated at 37 °C for 24 h. The tested compound (15µL, 50 µM) was then added and incubated at 37 °C for another 24 h. Then 20 µL of the sample was diluted to a final volume of 200 µL with 50 mM glycine–NaOH buffer (pH 8.0) containing

ACS Paragon Plus Environment

Page 68 of 91

Page 69 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

thioflavin T (5 µM). The detection method was the same as above. For the experiment of copper-induced Aβ fibrils disaggregation, the Aβ stock solution was diluted in 20 µM HEPES (pH 6.6) with 150 µM NaCl. The mixture of the peptide (10 µL, 25 µM, final concentration) with copper (10 µL, 25 µM, final concentration) was incubated at 37 oC for 24 h. The tested compound (10µL, 50 µM, final concentration) was then added and incubated at 37 °C for another 24 h. Then 20 µL of the sample was diluted to a final volume of 200 µL with 50 mM glycine–NaOH buffer (pH 8.0) containing thioflavin T (5 µM). The detection method was the same as above. TEM Assay For the metal-free experiment, Aβ stock solution was diluted with a 10 mM phosphate buffer (pH = 7.4); for the copper-induced experiment, Aβ stock solution was diluted with 20 µM HEPES (pH = 6.6) and 150 µM NaCl. The sample preparation was same as that for the ThT assay. Aliquots (10 µL) of the samples were placed on a carbon-coated copper/rhodium grid for 2 min. Each grid was stained with uranyl acetate (1%, 5 µL) for 2 min. After draining off the excess staining solution, the specimen was transferred for imaging in a transmission electron microscope (JEOL JEM-1400). All compounds are solubilized in the buffer which was used for the experiment. Metal-chelating Study The chelating studies were performed with a UV-Vis spectrophotometer. The absorption spectra of compound 8d (50 µM, final concentration) alone or in the

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

presence of CuSO4, FeSO4, FeCl3 or ZnCl2 (50 µM, final concentration) in buffer (20 mM HEPES, 150 mM NaCl, pH 7.4) were incubated for 30 min and then recorded at room temperature, respectively. For the stoichiometry of the compound-Cu2+ complex, a fixed amount of 8d (50 µM) was mixed with growing amounts of copper ion (0-100 µM), and the difference UV-vis spectra were examined to investigate the ratio of ligand/metal in the complex. Neuronal Cell Proliferation Assay16, 35 The undifferentiated rat neuronal cells PC12 (purchased from cell bank of the Chinese Academy of Sciences, Shanghai, China) were cultured in F12K medium supplemented with 10% horse serum, and 5% fetal bovine serum. The cells were placed into the wells of 6-well plates at 1 × 105 cells/well (used for morphological studies) and 96-well plates at 5 × 104 cells/mL (used for neuron numbers quantification) held in an incubator at 37 °C under a 5% CO2 atmosphere. After 24 h, the medium was replaced with fresh medium, and the undifferentiated neuronal cells were treated with vehicle (0.1% (v/v) DMSO), NGF (50 ng/mL, Peprotech, London, UK), or 0, 0.5, 1, 5, 10, 20, 40, or 50 µM of a quinoline-indol derivative for 3 days. At least six fields were randomly selected and photographed per well at 40 × magnification (morphological studies) and 4 × magnification (neuron numbers studies) using an EVOS FL microscope (EVOS FL Auto) equipped with a digital camera. Neurite length and neuron numbers were analyzed using Image-ProPlus based on morphological results. For the possible mechanism studies of mitogenic effect, neuronal survival was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl

ACS Paragon Plus Environment

Page 70 of 91

Page 71 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

tetrazolium bromide (MTT, 570 nm) assay and cells that were only treated with vehicle were set as control (100%). At least three independent experiments were performed. Anti-intracellular Oxidative Stress in SH-SY5Y cells SH-SY5Y cells were seeded at 1 × 104 cells/well in 96-well plates; After 24 h, the medium was removed and replaced with tested compounds at 37 °C and kept for another 24 h. The cells were washed with PBS and incubated with 5 µM of DCFH-DA (a fluorescent probe) in PBS at 37 °C in 5% CO2 for 30 min. After removal of DCFH-DA and further washing, the cells were exposed to 0.1 mM t-BuOOH (tert-butyl hydroperoxide, a compound used to induce oxidative stress) in PBS for 30 min. At the end of the incubation, the fluorescence of the cells from each well was measured (λ excitation = 485 nm, λ emission = 535 nm) with a multifunctional microplate reader (Molecular Devices, Flex Station 3). The antioxidant activity was expressed as percentage of control cells and calculated by the formula (Ft - Fnt) / (Ft’ - Fnt)×100, where Ft = absorbance of treated neurons with tested compound, Ft’ = absorbance of treated neurons without tested compound and Fnt = absorbance of neurons not treated with t-BuOOH. Metabolic Stability The rat liver microsomes were prepared according to the previously described methods.31,

48

Compounds 7g•HCl, 8c•HCl, 8d•HCl, 8g•HCl, CQ, donepezil and

testosterone were dissolved in CH3CN as 10 mM stock solution and incubated with rat liver microsomes (1 mg of protein per mL, final concentration) at a final

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

concentration of 100 µM in a final volume of 0.5 mL buffer solution (PBS, 100.0 mM; MgCl2, 3.0 mM; 1.3 mM β-NADPNa2, 3.3 mM glucose 6-phosphate; 0.4 Units/mL glucose 6-phosphate dehydrogenase; pH = 7.4). The samples were incubated for various time intervals (0, 5, 10, 30, 60, 90, 120, 150, 180 min) at 37 °C in a water bath. The incubation were terminated at different time points by adding 0.5 mL of ice-cold CH3CN. A parallel incubation was performed in the absent of NADPH-regenerating system and microsomes as the negative control, and the reactions were terminated after the corresponding time incubation. After centrifugation with 12500 rpm at 4 °C for 10 min, the supernatant were directly analyzed by the HPLC-UV systems (Agilent HPLC 1200 instrument). Three independent experiments were performed in triplicate. Acute Toxicity and Histological Studies The procedures for acute toxicity study were followed the similar protocols in our previous studies.31, 49 C57BL/6 mice (male, 12 weeks old, 25-30 g) purchased from the laboratory animal center of Sun Yat-sen University (Guangzhou, China) were used to evaluate the acute toxicity of compounds 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl. Mice were maintained with a 12-h light/dark cycle (light from 07:00 to 19:00) at 20-22 °C with a 60-70% relative humidity. Sterile food and water were provided according to institutional guidelines. Prior to each experiment, mice were fasted overnight and allowed free access to water. Compound 7g•HCl, 8c•HCl, 8d•HCl and 8g•HCl was suspension in 0.5% carboxymethyl cellulose sodium (CMC-Na) salt solution (677, 1333, and 2000 mg/kg) and given via oral administration according to the divided experimental groups. After the administration of the compounds, the mice

ACS Paragon Plus Environment

Page 72 of 91

Page 73 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

were observed continuously for the first 4 h for any abnormal behavior and mortality changes, intermittently for the next 24 h, and occasionally thereafter for 14 days for the onset of any delayed effects. All animals were sacrificed on the 14th day after drug administration and were macroscopically examined for possible damage to the heart, liver, lung, and brain. The organs were carefully collected, post-fixed in 4% paraformaldehyde for 24 h, and then embedded in paraffin. For the brain, coronal sections (4 µm) were cut through the entire hippocampus, for other organs, the sections (4 µm) were cut through the maximum transverse sections and used for HE staining. The paraffin sections were conventionally dewaxed and rehydrated, followed by hematoxylin staining for 5 min and eosin staining for 1 min. Then, the sections were dehydrated in a graded series of alcohol, made transparent with xylene, and sealed. The histopathological abnormalities were investigated under EVOS FL microscope (EVOS FL Auto) equipped with a digital camera. In Living Adult C57BL/6 Mice Hippocampal Cell Proliferation17, 25, 29 Adult (12 weeks old) wild type C57BL/6 mice were purchased from Guangdong Medical Laboratory Animal Center (Guangdong, China) for in vivo study. In order to ensure a low baseline level of neurogenesis, mice were individually housed without access to running wheels beginning 7 days prior to pump implantation and throughout the entire procedure, as both social activity and voluntary exercise could stimulated hippocampal neurogenesis. All experiments were strictly executed in accordance with the Guide for the Care and Use of Laboratory Animals, and were approved by the Animal Experimentation Ethics Committee of Sun Yat-sen University. For the ICV

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

method group, each compounds were dissolved at 10 µM (containing 0.05% DMSO) in an artificial cerebrospinal fluid (aCSF: NaCl (128 mM), KCl (2.5 mM), CaCl2 (0.95 mM), MgCl2 (1.9 mM)), and then infused ICV into the left lateral ventricle of six adult (12 weeks old) wild type C57BL/6 mice by means of surgically implanted Alzet osmotic minipumps which delivered the solution into animals at a constant rate of 0.5 µL/h for 7 days, respectively. Surgical implantation of Alzet osmotic minipumps (Alzet 1007D; Cupertino, CA) was conducted as follows. Animals were anesthetized with intraperitoneally administered sodium pentobarbital (dosage, 0.15 mL/10g; concentration, 0.3% in normal saline), and the back of the neck and upper back area were then shaved and swabbed with 70% ethanol and betadine. A small midline incision was made in the skin between the scapulae to allow use of a hemostat to create a small pocket by spreading the subcutaneous connective tissue apart. The pump was inserted into the pocket with the flow moderator pointing away from the incision, and the incision was closed with surgical thread. Surgery to implant the cannula into the lateral ventricle followed immediately, while animals were still anesthetized. The top portion of the head was shaved and the animal was then placed in a stereotactic holder. The head was swabbed with 70% alcohol followed by betadine, and an incision was made in the scalp to expose the surface of the skull in the area of bregma. The skull was cleaned of fascial tissue and a burr hole was made in the skull such that the guide cannula could be lowered into the burr hole to a depth H = + 3 mm deep to the pial surface, AP = - 0.3 mm anteroposterior relative to bregma, and L = - 1.3 mm lateral to midline. The guide cannula was secured to the

ACS Paragon Plus Environment

Page 74 of 91

Page 75 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

skull with dental cement and the scalp incision was closed by suture thread. Animals were then placed in a clean warm cage on a heating pad until mobile. For the oral administration group, the mice in each group were administered the corresponding drug dosage in a 0.5% CMC-Na solution (0.02 mL/g) by intragastric infusion for 7 days (using a blank 0.5% CMC-Na solution as placebo for vehicle groups). All mice had free access to food and water, bromodeoxyuridine (BrdU, Sigma-Aldrich) was injected intraperitoneally at 50 mg/kg/day for six days during pump infusion in order to detect agents that either promote proliferation or augment survival of proliferating neural stem cells in the hippocampus. The injection site was alternated between right and left sides. Animals were monitored daily for general health and weighed every day. Accumulated weight loss > 20% relative to pre-surgical weight was considered a sign of sickness and any such animals were euthanized for humane treatment. Twenty-four hours after the final BrdU administration, mice were sacrificed by transcardial perfusion with 4 % paraformaldehyde at pH 7.4, and their brains were processed for immunohistochemical detection of incorporated BrdU in the hippocampus. Dissected brains were immersed in 4 % paraformaldehyde overnight at 4 °C, then embedded in paraffin before being sectioned with a Leica SM2000R sliding microtome coronally into 30 µM thick free-floating sections. Unmasking of BrdU antigen was achieved through incubating tissue sections for two hours in 50% formamide / 2X SSC at 65 °C, followed by five minute wash in 2X SSC and subsequent incubation for 30 minutes in 2 M HCl at 37 °C. Sections were processed for immunohistochemical staining with mouse monoclonal anti-BrdU (1:100,

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Sigma-Aldrich). Images were analyzed with a motorized research microscope, the number of BrdU+ cells in the entire hippocampus in the contralateral hemisphere (opposite side of surgically implanted pump) was quantified by counting BrdU+ cells within hippocampus in every fifth section throughout the entire hippocampus and then normalizing for hippocampus volume. Pharmacokinetics Analysis and Log BB Values Studies Pharmacokinetic properties of 8d•HCl were analyzed by HDBioscieces and Medicilon Companies, Shanghai, China. Male SD rats with body weight of 220-250 g, which were purchased from Shanghai SIPPR-BK LAB Animal Ltd., Shanghai, China. Compound 8d•HCl was dissolved in 5% DMSO, 10% Solutol and 85% saline for intravenous administration (IV, 1 mg/mL) and oral administration (PO, 3 mg/mL). A single dosage of 5 mg/kg for IV (three SD rats) and 30 mg/kg for PO (three SD rats) of the formulated compounds were administrated, with the blood samples were taken at 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h, 8 h, and 24 h after the compound administration. The concentration of the corresponding compound in blood were analyzed by LC-MS/MS (Shimadzu liquid chromatographic system and API4000 mass spectrometer, Applied Biosystems, Ontario, Canada). For the log BB values studies, a single dosage of 30 mg/kg for PO (three SD rats) of the formulated compounds were administrated, 1 h after the oral administration of compound 8d•HCl, the three SD rats were sacrificed, the plasma, brains were collected from the three animals, the concentration of the corresponding compound in blood and brain were analyzed by LC-MS/MS.

ACS Paragon Plus Environment

Page 76 of 91

Page 77 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Cognitive and Memory Improvement in APP/PS1 AD Mice by Long-term Oral Uptake of Compounds31 Animals. Six-month-old double transgenic (APP/PS1) AD mice and C57BL/6 littermates mice (wild type, WT) were purchased from the Beijing HFK Bioscience Co., Ltd (Beijing, P. R. China), the APP/PS1 mice were randomly allocated into 4 groups (n = 10 for each group): vehicle, CQ (30 mg/kg/day), 8d•HCl (30 mg/kg/day) and 8g•HCl (30 mg/kg/day). The mice were maintained on a 12 h light/dark cycle (light from 07:00 to 19:00) at 23 °C and 60-70% relative humidity, with ad libitum access to sterile food and water. All experiments were strictly executed in accordance with the Guide for the Care and Use of Laboratory Animals and were approved by the Animal Experimentation Ethics Committee of Sun Yat-sen University. The animals were taken to their individual cages and the mice in each group were administered the corresponding drug dosage in a 0.5% CMC-Na solution (0.02 mL/g) by intragastric infusion throughout the entire assay (using a blank 0.5% CMC-Na solution as placebo for vehicle and WT groups). Morris Water Maze Test. Seventy-four days later, behavioral performance was evaluated using the Morris water maze task. (The water maze apparatus consisted of a circular pool, 65 cm in diameter and 40 cm high, filled to a depth of 17 cm with fresh water (kept at 25 °C, the fresh water was refilled each day before the trails), which was made opaque by the addition of non-toxic white TiO2 at a concentration of 0.25 g/L.) The task demanded incremental learning of the location of a fixed, hidden platform (6.5 cm in diameter and 16 cm high, submerged 1.0 cm under the opaque

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

water level) throughout the training period. In the single training assay, each mouse underwent reference memory training (four trials per day and a 30 min intertrial interval) with a fixed hidden-platform located in the center of third quadrant (southwest) of the pool for 5 consecutive days. The mouse were placed in the water facing the wall of the pool from one of the four designated starting locations (west, north, south and east), each of the starting locations was used once in the four trials and the order of the starting locations was varied between the 5 consecutive training days. If the mouse did not find the platform within 90 s, the mouse was guided onto the platform and the escape latency was recorded as 90 s. For each trial, the mouse was allowed 15 s on the platform, and the escape latency and trajectories were recorded using a computerized-digitizing video-tracking system (Chromo track, Meng Tai Instruments, ChengDu, China). All of experiments were performed between 9:00 and 15:00 at 2 h after each drug infusion, and the room temperature was maintained at 25 °C. The extra-maze visual cues remained in fixed positions in the room where the Morris water maze apparatus was located; olfactory auditory cues were minimized by maintaining a fresh and quiet environment. To assess memory retention, a spatial probe trial was performed 24 h after the last training trial, with the platform removed from the pool and the two-fold diameter of the original platform location was set as the effective region. Each mouse was allowed to swim freely in the pool for 90 s (only one trial) from the northeast, a new starting location, which was farthest from the virtual platform location. The number of times the swimming mouse crossed the virtual platform location (original platform location), the average swimming speed,

ACS Paragon Plus Environment

Page 78 of 91

Page 79 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

the swimming path length as well as time spent the in the virtual platform, the effective

region

and

the

correct

quadrant

were

recorded

by

the

computerized-digitizing video-tracking system and to analyze the level of memory retention. Aβ Plaque Immunohistochemistry in the Brain of APP/PS1 AD Mice After the Morris water maze experiment, animals were deeply anesthetized with an intraperitoneal injection of and transcardially perfused with 4% paraformaldehyde. The brain was carefully removed, post-fixed in 4% paraformaldehyde for 24 h, and then embedded in paraffin. Three coronal sections (30 µm in thickness and 120-µm interval) were cut through the medial hippocampus for immunohistochemical staining. The sections were pretreated with 3% H2O2 for 25 min to block endogenous peroxidase activity, then the sections washed with distilled water and 0.1 M Tris-buffered saline (TBS), PH 7.4. Sections were immunostained using the following antibodies: incubated at 4 °C overnight with the primary amyloid-β monoclonal antibody (mOC64; 1:300; Abcam, UK), subsequently rinsed and incubated with biotinylated secondary antibody (Dako) for 50 min, finally with the chromogen diaminobenzidine. After staining, the sections were dehydrated and observed under EVOS FL microscope (EVOS FL Auto) equipped with a digital camera. The Aβ plaque deposition in brain was calculated as the percentage of the Aβ deposition area with respect to the overall area in three representative images for each mice by using the ImageJ software. Statistical Analysis

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 80 of 91

All data were expressed as mean ± standard deviation, results were subjected to Student’s t test or one-way analysis of variance (ANOVA) followed by Dunnett’s test. P < 0.05 was accepted to indicate the significance. Supporting Information Supporting Information Available: The HPLC data of the target compounds and the method for PAMPA Corresponding Author *For

L.H.:

phone,

+086-20-3994-3051;

[email protected].

For

X.L.:

fax,

+086-20-3994-3051;

phone,

+086-20-3994-3050;

e-mail, fax,

+086-20-3994-3050; e-mail, [email protected]. Acknowledgments We thank the National Natural Science Foundation of China (No. 21772241, No. 21302235), Guangdong Natural Science Foundation (2014A030313124), Guangdong High-level personnel of special support program -Young top-notch talent project (2015TQ01R244), Guangzhou Pearl River New Star Fund Science and Technology Planning Project (201610010111) and The Fundamental Research Funds for the Central Universities (15ykpy04) for financial support of this study. Abbreviations Aβ, amyloid-β; AD, Alzheimer's disease; ADME, absorption, distribution, metabolism, and excretion; aCSF, artificial cerebral spinal fluid; ALS, amyotrophic lateral sclerosis; BBB, blood-brain barrier; BrdU, bromodeoxyuridine; CA, cornus ammonis, CMC-Na, carboxymethyl cellulose sodium; CNS, central nervous system;

ACS Paragon Plus Environment

Page 81 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

CQ, clioquinol; CSF, cerebrospinal fluid; DCFH-DA, dichlorofluorescein diacetate; DG, dentate gyrus; ERK, extracellular signal-regulated kinase; HD, Huntington’s disease; HE, hematoxylin and eosin; HRMS, high-resolution mass spectra; ICV, intracerebroventricular; IP, intraperitoneal; MAPKs, mitogen-activated protein kinase; MTDLs,

multi-target-directed

ligands;

MTT,

3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide; MWM, Morris water maze; NMDAR, N-methyl-D-aspartate (NMDA) receptor; ORAC, oxygen radical Absorbance Capacity; PAMPA, parallel artificial membrane permeability; PBS, phosphate buffer; PD, Parkinson’s disease; PO, oral administration; ROS, reactive oxygen species; SAR, structure-activity relationship; SGZ, subgranular zone; TBI, traumatic brain injury; t-BuOOH, tert-butyl hydroperoxide; TEM, transmission electron microscopy; ThT, thioflavin T; TLC, thin-layer chromatography; WT, wide type. REFERENCE 1.

Prince, M.; Comas-Herrera, A.; Knapp, M.; Guerchet, M.; Karagiannidou, M.

World Alzheimer Report 2016. Improving Healthcare for People Living with Dementia: Coverage, Quality and Costs Now and in the Future. Alzheimer's Disease International: London 2016, pp1-131. 2.

Amiri, H.; Saeidi, K.; Borhani, P.; Manafirad, A.; Ghavami, M.; Zerbi, V.

Alzheimer's disease: pathophysiology and applications of magnetic nanoparticles as MRI theranostic agents. ACS Chem. Neurosci. 2013, 4, 1417-1429. 3.

Sola, I.; Aso, E.; Frattini, D.; Lopez-Gonzalez, I.; Espargaro, A.; Sabate, R.; Di

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Pietro, O.; Luque, F. J.; Clos, M. V.; Ferrer, I.; Munoz-Torrero, D. Novel levetiracetam derivatives that are effective against the Alzheimer-like phenotype in mice: synthesis, in vitro, ex vivo, and in vivo efficacy studies. J. Med. Chem. 2015, 58, 6018-6032. 4.

Farina, R.; Pisani, L.; Catto, M.; Nicolotti, O.; Gadaleta, D.; Denora, N.;

Soto-Otero, R.; Mendez-Alvarez, E.; Passos, C. S.; Muncipinto, G.; Altomare, C. D.; Nurisso, A.; Carrupt, P. A.; Carotti, A. Structure-based design and optimization of multitarget-directed 2H-chromen-2-one derivatives as potent inhibitors of monoamine oxidase B and cholinesterases. J. Med. Chem. 2015, 58, 5561-5578. 5.

Viayna, E.; Sola, I.; Bartolini, M.; De Simone, A.; Tapia-Rojas, C.; Serrano, F. G.;

Sabate, R.; Juarez-Jimenez, J.; Perez, B.; Luque, F. J.; Andrisano, V.; Clos, M. V.; Inestrosa, N. C.; Munoz-Torrero, D. Synthesis and multitarget biological profiling of a novel family of rhein derivatives as disease-modifying anti-Alzheimer agents. J. Med. Chem. 2014, 57, 2549-2567. 6.

Lopez-Iglesias, B.; Perez, C.; Morales-Garcia, J. A.; Alonso-Gil, S.;

Perez-Castillo, A.; Romero, A.; Lopez, M. G.; Villarroya, M.; Conde, S.; Rodriguez-Franco, M. I. New melatonin-N,N-dibenzyl(N-methyl)amine hybrids: potent neurogenic agents with antioxidant, cholinergic, and neuroprotective properties as innovative drugs for Alzheimer's disease. J. Med. Chem. 2014, 57, 3773-3785. 7.

Jones, M. R.; Mathieu, E.; Dyrager, C.; Faissner, S.; Vaillancourt, Z.; Korshavn,

K. J.; Lim, M. H.; Ramamoorthy, A.; Wee Yong, V.; Tsutsui, S.; Stys, P. K.; Storr, T. Multi-target-directed phenol-triazole ligands as therapeutic agents for Alzheimer's

ACS Paragon Plus Environment

Page 82 of 91

Page 83 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

disease. Chem. Sci. 2017, 8, 5636-5643. 8.

Mujika, J. I.; Rodriguez-Guerra Pedregal, J.; Lopez, X.; Ugalde, J. M.;

Rodriguez-Santiago, L.; Sodupe, M.; Marechal, J.-D. Elucidating the 3D structures of Al(iii)-Aβ complexes: a template free strategy based on the pre-organization hypothesis. Chem. Sci. 2017, 8, 5041-5049. 9.

Barnham, K. J.; Bush, A. I. Biological metals and metal-targeting compounds in

major neurodegenerative diseases. Chem. Soc. Rev. 2014, 43, 6727-6749. 10. Savelieff, M. G.; DeToma, A. S.; Derrick, J. S.; Lim, M. H. The ongoing search for small molecules to study metal-associated amyloid-beta species in Alzheimer's disease. Acc. Chem. Res. 2014, 47, 2475-2482. 11. Robert, A.; Liu, Y.; Nguyen, M.; Meunier, B. Regulation of copper and iron homeostasis by metal chelators: a possible chemotherapy for Alzheimer’s disease. Acc. Chem. Res. 2015, 48, 1332-1339. 12. Faux, N. G.; Ritchie, C. W.; Gunn, A.; Rembach, A.; Tsatsanis, A.; Bedo, J.; Harrison, J.; Lannfelt, L.; Blennow, K.; Zetterberg, H.; Ingelsson, M.; Masters, C. L.; Tanzi, R. E.; Cummings, J. L.; Herd, C. M.; Bush, A. I. PBT2 rapidly improves cognition in Alzheimer's Disease: additional phase II analyses. J. Alzheimer's Dis. 2010, 20, 509-516. 13. Adlard, P. A.; Cherny, R. A.; Finkelstein, D. I.; Gautier, E.; Robb, E.; Cortes, M.; Volitakis, I.; Liu, X.; Smith, J. P.; Perez, K.; Laughton, K.; Li, Q.-X.; Charman, S. A.; Nicolazzo, J. A.; Wilkins, S.; Deleva, K.; Lynch, T.; Kok, G.; Ritchie, C. W.; Tanzi, R. E.; Cappai, R.; Masters, C. L.; Barnham, K. J.; Bush, A. I. Rapid restoration of

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

cognition in Alzheimer's transgenic mice with 8-hydroxy quinoline analogs is associated with decreased interstitial Aβ. Neuron 2008, 59, 43-55. 14. Mangialasche, F.; Solomon, A.; Winblad, B.; Mecocci, P.; Kivipelto, M. Alzheimer's disease: clinical trials and drug development. Lancet Neurol. 2010, 9, 702-716. 15. Beck, M. W.; Oh, S. B.; Kerr, R. A.; Lee, H. J.; Kim, S. H.; Kim, S.; Jang, M.; Ruotolo, B. T.; Lee, J.-Y.; Lim, M. H. A rationally designed small molecule for identifying an in vivo link between metal-amyloid-β complexes and the pathogenesis of Alzheimer's disease. Chem. Sci. 2015, 6, 1879-1886. 16. Chang, P. T.; Talekar, R. S.; Kung, F. L.; Chern, T. R.; Huang, C. W.; Ye, Q. Q.; Yang, M. Y.; Yu, C. W.; Lai, S. Y.; Deore, R. R.; Lin, J. H.; Chen, C. S.; Chen, G. S.; Chern, J. W. A newly designed molecule J2326 for Alzheimer's disease disaggregates amyloid fibrils and induces neurite outgrowth. Neuropharmacology 2015, 92, 146-157. 17. Pieper, A. A.; Xie, S.; Capota, E.; Estill, S. J.; Zhong, J.; Long, J. M.; Becker, G. L.; Huntington, P.; Goldman, S. E.; Shen, C. H.; Capota, M.; Britt, J. K.; Kotti, T.; Ure, K.; Brat, D. J.; Williams, N. S.; MacMillan, K. S.; Naidoo, J.; Melito, L.; Hsieh, J.; De Brabander, J.; Ready, J. M.; McKnight, S. L. Discovery of a proneurogenic, neuroprotective chemical. Cell 2010, 142, 39-51. 18. Pieper, A. A.; McKnight, S. L.; Ready, J. M. P7C3 and an unbiased approach to drug discovery for neurodegenerative diseases. Chem. Soc. Rev. 2014, 43, 6716-6726. 19. Davies, S. G.; Kennewell, P. D.; Russell, A. J.; Seden, P. T.; Westwood, R.;

ACS Paragon Plus Environment

Page 84 of 91

Page 85 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Wynne, G. M. Stemistry: the control of stem cells in situ using chemistry. J. Med. Chem. 2015, 58, 2863-2894. 20. Marschallinger, J.; Schaffner, I.; Klein, B.; Gelfert, R.; Rivera, F. J.; Illes, S.; Grassner, L.; Janssen, M.; Rotheneichner, P.; Schmuckermair, C.; Coras, R.; Boccazzi, M.; Chishty, M.; Lagler, F. B.; Renic, M.; Bauer, H.-C.; Singewald, N.; Blumcke, I.; Bogdahn, U.; Couillard-Despres, S.; Lie, D. C.; Abbracchio, M. P.; Aigner, L. Structural and functional rejuvenation of the aged brain by an approved anti-asthmatic drug. Nat. Commun. 2015, 6, 8466. 21. Encinas, J. M.; Vaahtokari, A.; Enikolopov, G. Fluoxetine targets early progenitor cells in the adult brain. Proc. Natl. Acad. Sci. U. S. A. 2006, 103, 8233-8238. 22. Wang, J. M.; Singh, C.; Liu, L.; Irwin, R. W.; Chen, S.; Chung, E. J.; Thompson, R. F.; Brinton, R. D. Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer's disease. Proc. Natl. Acad. Sci. U. S. A. 2010, 107, 6498-6503. 23. Teo, J. L.; Ma, H.; Nguyen, C.; Lam, C.; Kahn, M. Specific inhibition of CBP/beta-catenin interaction rescues defects in neuronal differentiation caused by a presenilin-1 mutation. Proc. Natl. Acad. Sci. U. S. A. 2005, 102, 12171-12176. 24. Deshmukh, V. A.; Tardif, V.; Lyssiotis, C. A.; Green, C. C.; Kerman, B.; Kim, H. J.; Padmanabhan, K.; Swoboda, J. G.; Ahmad, I.; Kondo, T.; Gage, F. H.; Theofilopoulos, A. N.; Lawson, B. R.; Schultz, P. G.; Lairson, L. L. A regenerative approach to the treatment of multiple sclerosis. Nature 2013, 502, 327-332. 25. Wang, G.; Han, T.; Nijhawan, D.; Theodoropoulos, P.; Naidoo, J.; Yadavalli, S.;

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 86 of 91

Mirzaei, H.; Pieper, A. A.; Ready, J. M.; McKnight, S. L. P7C3 neuroprotective chemicals function by activating the rate-limiting enzyme in NAD salvage. Cell 2014, 158, 1324-1334. 26. Naidoo, J.; De Jesus-Cortes, H.; Huntington, P.; Estill, S.; Morlock, L. K.; Starwalt, R.; Mangano, T. J.; Williams, N. S.; Pieper, A. A.; Ready, J. M. Discovery of a

neuroprotective

chemical,

(S)-N-(3-(3,6-dibromo-9H-carbazol-9-yl)-2-fluoropropyl)-6-methoxypyridin-2-amine [(-)-P7C3-S243], with improved druglike properties. J. Med. Chem. 2014, 57, 3746-3754. 27. Tesla, R.; Wolf, H. P.; Xu, P.; Drawbridge, J.; Estill, S. J.; Huntington, P.; McDaniel, L.; Knobbe, W.; Burket, A.; Tran, S.; Starwalt, R.; Morlock, L.; Naidoo, J.; Williams, N. S.; Ready, J. M.; McKnight, S. L.; Pieper, A. A. Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 17016-17021. 28. De Jesús-Cortés, H.; Xu, P.; Drawbridge, J.; Estill, S. J.; Huntington, P.; Tran, S.; Britt, J.; Tesla, R.; Morlock, L.; Naidoo, J.; Melito, L. M.; Wang, G.; Williams, N. S.; Ready, J. M.; McKnight, S. L.; Pieper, A. A. Neuroprotective efficacy of aminopropyl carbazoles in a mouse model of Parkinson disease. Proc. Natl. Acad. Sci. U. S. A. 2012, 109, 17010-17015. 29. MacMillan, K. S.; Naidoo, J.; Liang, J.; Melito, L.; Williams, N. S.; Morlock, L.; Huntington, P. J.; Estill, S. J.; Longgood, J.; Becker, G. L.; McKnight, S. L.; Pieper, A. A.; De Brabander, J. K.; Ready, J. M. Development of proneurogenic, neuroprotective

ACS Paragon Plus Environment

Page 87 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

small molecules. J. Am. Chem. Soc. 2011, 133, 1428-1437. 30. Ramirez-Rodriguez, G.; Klempin, F.; Babu, H.; Benitez-King, G.; Kempermann, G. Melatonin modulates cell survival of new neurons in the hippocampus of adult mice. Neuropsychopharmacology 2009, 34, 2180-2191. 31. Wang, Z.; Wang, Y.; Wang, B.; Li, W.; Huang, L.; Li, X. Design, synthesis, and evaluation of orally available clioquinol-moracin M hybrids as multitarget-directed ligands for cognitive improvement in a rat model of neurodegeneration in Alzheimer’s disease. J. Med. Chem. 2015, 58, 8616-8637. 32. DeToma, A. S.; Krishnamoorthy, J.; Nam, Y.; Lee, H. J.; Brender, J. R.; Kochi, A.; Lee, D.; Onnis, V.; Congiu, C.; Manfredini, S.; Vertuani, S.; Balboni, G.; Ramamoorthy,

A.;

Lim,

M.

H.

Interaction

and

reactivity

of

synthetic

aminoisoflavones with metal-free and metal-associated amyloid-beta. Chem. Sci. 2014, 5, 4851-4862. 33. Porter, M. R.; Kochi, A.; Karty, J. A.; Lim, M. H.; Zaleski, J. M. Chelation-induced diradical formation as an approach to modulation of the amyloid-beta aggregation pathway. Chem. Sci. 2015, 6, 1018-1026. 34. Derrick, J. S.; Kerr, R. A.; Nam, Y.; Oh, S. B.; Lee, H. J.; Earnest, K. G.; Suh, N.; Peck, K. L.; Ozbil, M.; Korshavn, K. J.; Ramamoorthy, A.; Prabhakar, R.; Merino, E. J.; Shearer, J.; Lee, J.-Y.; Ruotolo, B. T.; Lim, M. H. A redox-active, compact molecule for cross-linking amyloidogenic peptides into nontoxic, off-pathway aggregates: in vitro and in vivo efficacy and molecular mechanisms. J. Am. Chem. Soc. 2015, 137, 14785-14797.

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

35. Yu, C. W.; Chang, P. T.; Hsin, L. W.; Chern, J. W. Quinazolin-4-one derivatives as selective histone deacetylase-6 inhibitors for the treatment of Alzheimer's disease. J Med. Chem. 2013, 56, 6775-6791. 36. Lambeng, N.; Willaime-Morawek, S.; Mariani, J.; Ruberg, M.; Brugg, B. Activation of mitogen-activated protein kinase pathways during the death of PC12 cells is dependent on the state of differentiation. Mol. Brain Res. 2003, 111, 52-60. 37. Liew, K. F.; Chan, K. L.; Lee, C. Y. Blood-brain barrier permeable anticholinesterase aurones: synthesis, structure-activity relationship, and drug-like properties. Eur. J. Med. Chem. 2015, 94, 195-210. 38. Shao, Y. X.; Huang, M.; Cui, W.; Feng, L. J.; Wu, Y.; Cai, Y.; Li, Z.; Zhu, X.; Liu, P.; Wan, Y.; Ke, H.; Luo, H. B. Discovery of a phosphodiesterase 9A inhibitor as a potential hypoglycemic agent. J. Med. Chem. 2014, 57, 10304-10313. 39. Rankovic, Z. CNS drug design: balancing physicochemical properties for optimal brain exposure. J. Med. Chem. 2015, 58, 2584-2608. 40. Rankovic, Z. CNS physicochemical property space shaped by a diverse set of molecules with experimentally determined exposure in the mouse brain. J. Med. Chem. 2017, 60, 5943-5954. 41. Zheng, H.; Weiner, L. M.; Bar-Am, O.; Epsztejn, S.; Cabantchik, Z. I.; Warshawsky, A.; Youdim, M. B. H.; Fridkin, M. Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer’s, Parkinson’s, and other neurodegenerative diseases. Bioorg. Med. Chem. 2005, 13, 773-783.

ACS Paragon Plus Environment

Page 88 of 91

Page 89 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

42. Rodríguez-Franco,

M.

I.;

Fernández-Bachiller,

M.

I.;

Pérez,

C.;

Hernández-Ledesma, B.; Bartolomé, B. Novel tacrine−melatonin hybrids as dual-acting drugs for Alzheimer disease, with improved acetylcholinesterase inhibitory and antioxidant properties. J. Med. Chem. 2006, 49, 459-462. 43. Decker, M.; Kraus, B.; Heilmann, J. Design, synthesis and pharmacological evaluation of hybrid molecules out of quinazolinimines and lipoic acid lead to highly potent and selective butyrylcholinesterase inhibitors with antioxidant properties. Bioorg. Med. Chem. 2008, 16, 4252-4261. 44. Di, L.; Kerns, E. H.; Fan, K.; McConnell, O. J.; Carter, G. T. High throughput artificial membrane permeability assay for blood–brain barrier. Eur. J. Med. Chem. 2003, 38, 223-232. 45. Camps, P.; Formosa, X.; Galdeano, C.; Muñoz-Torrero, D.; Ramírez, L.; Gómez, E.; Isambert, N.; Lavilla, R.; Badia, A.; Clos, M. V.; Bartolini, M.; Mancini, F.; Andrisano, V.; Arce, M. P.; Rodríguez-Franco, M. I.; Huertas, Ó.; Dafni, T.; Luque, F. J.

Pyrano[3,2-c]quinoline−6-chlorotacrine

hybrids

as

a

novel

family

of

acetylcholinesterase- and β-amyloid-directed anti-Alzheimer compounds. J. Med. Chem. 2009, 52, 5365-5379. 46. Wohnsland, F.; Faller, B. High-throughput permeability pH profile and high-throughput alkane/water log P with artificial membranes. J. Med. Chem. 2001, 44, 923-930. 47. Rosini, M.; Simoni, E.; Bartolini, M.; Cavalli, A.; Ceccarini, L.; Pascu, N.; McClymont, D. W.; Tarozzi, A.; Bolognesi, M. L.; Minarini, A.; Tumiatti, V.;

ACS Paragon Plus Environment

Journal of Medicinal Chemistry 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Andrisano, V.; Mellor, I. R.; Melchiorre, C. Inhibition of acetylcholinesterase, β-amyloid aggregation, and NMDA receptors in Alzheimer’s disease: a promising direction for the multi-target-directed ligands gold rush. J. Med. Chem. 2008, 51, 4381-4384. 48. Brunschweiger, A.; Iqbal, J.; Umbach, F.; Scheiff, A. B.; Munkonda, M. N.; Sevigny, J.; Knowles, A. F.; Muller, C. E. Selective nucleoside triphosphate diphosphohydrolase-2 (NTPDase2) inhibitors: nucleotide mimetics derived from uridine-5'-carboxamide. J. Med. Chem. 2008, 51, 4518-4528. 49. Wang, Z.; Wang, Y.; Li, W.; Liu, Z.; Luo, Z.; Sun, Y.; Wu, R.; Huang, L.; Li, X. Computer-assisted designed "selenoxy-chinolin": a new catalytic mechanism of the GPx-like cycle and inhibition of metal-free and metal-associated Aβ aggregation. Dalton Trans. 2015, 44, 20913-20925.

ACS Paragon Plus Environment

Page 90 of 91

Page 91 of 91 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of Medicinal Chemistry

Table of Contents Graphic

ACS Paragon Plus Environment