the development of biologically active compounds and ... · 생체활성물질 개발 및 방향족...

136
理學博士學位 請求論文 생체활성물질 개발 및 방향족 친전자성 첨가반응에 관한 고찰 The Development of Biologically Active Compounds and Electrophilic Aromatic Addition Reaction 20052指導敎授 池 大 潤 이 논문을 박사학위 논문으로 제출함 Inha University Chemistry (Organic Chemistry) Ekaruth Srisook

Upload: others

Post on 19-Feb-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

  • 理學博士學位 請求論文

    생체활성물질 개발 및 방향족 친전자성

    첨가반응에 관한 고찰

    The Development of Biologically Active Compounds

    and Electrophilic Aromatic Addition Reaction

    2005년 2월

    指導敎授 池 大 潤

    이 논문을 박사학위 논문으로 제출함

    Inha University

    Chemistry (Organic Chemistry)

    Ekaruth Srisook

  • 理學博士學位 請求論文

    생체활성물질 개발 및 방향족 친전자성

    첨가반응에 관한 고찰

    The Development of Biologically Active Compounds

    and Electrophilic Aromatic Addition Reaction

    2005년 2월

    Inha University

    Chemistry (Organic Chemistry)

    Ekaruth Srisook

  • Contents

    • Abstract (English) ….. 6

    • Abstract (Korean) ….. 8 • Part 1. The Syntheses of 3-Substituted 4-(Pyridin-2-ylthio)indoles

    via Leimgruber-Batcho Indole Synthesis ….. 10

    Introduction …...11

    Results and discussion ….. 22

    Conclusions ….. 27

    Experimental section ….. 28

    References ….. 35

    NMR spectra ….. 39

    • Part 2. Structural Modification of Nitric Oxide Inhibitors ….. 50

    Introduction ….. 51

    Chemistry ….. 56

    Biological results and discussion ….. 59

    Conclusions ….. 62

    Experimental section ….. 63

    References ….. 70

    NMR spectra ….. 74

    • Part 3. Electrophilic Aromatic Addition Reaction: AdEAr ….. 84

    Introduction ….. 85

    Results and discussion ….. 91

    1. Synthesis of precursors ….. 91

  • 2. AdEAr reaction of 1-methoxynaphalene ….. 93 3. Reaction of 8-methoxyquinaldine under various conditions

    ….. 95

    4. AdEAr reaction of some selected compounds ….. 97 5. Mechanistic studies ….. 99 6. Reaction and application of addition product …..102

    Conclusions …..104

    Experimental section …..105

    References …..113

    NMR spectra …..115

    Acknowledgement …..137

  • 6

    Abstract

    Development of Biologically Active Compounds and Electrophilic

    Aromatic Addition Reaction

    Compounds which are strongly binding to serotonin transporter or nitric

    oxide synthases (NOS) would be new radiopharmaceuticals as serotonin

    selective reuptake inhibitors (SSRIs) or NOS inhibitors, respectively.

    Based on the structural diversities of selective serotonin reuptake

    inhibitors recently published, new family of ligands, 3-(amino- and

    hydroxymethyl)-4-(5-iodopyridin-2-ylthio)indoles have been designed for

    SSRI. These target compounds have been designed by a combination of

    characteristically distinct moieties proven to impart successful binding

    ability. The syntheses of 3-substituted 4-(5-iodopyridin-2-ylthio)indoles

    are described. Key intermediate 1-(5-iodopyridin-2-ylthio)-2-methyl-3-

    nitrobenzene (6) was achieved by nucleophilic aromatic substitution of

    chloropyridine 7 with thiophenol 8. A modified Leimgruber-Batcho indole

    synthesis from 1-(5-iodopyridin-2-ylthio)-2-methyl-3-nitrobenzene was

    used as a key step of this synthetic route. Unfortunately, these two

    compounds as well as other derivatives showed low binding affinities

    toward serotonin transporter. Nitric oxide (NO) or nitrogen monoxide, a

    small free radical, is generated by nitric oxide synthases (NOS). The

    overproductions of NO from iNOS, the inducible isoform of NOS, have

    been implicated in the pathophysiology of ischemic neuronal death and

    NOS inhibitors protected neurons in these animal models. A study of the

    regulation of NO overproductions is important because it prevents cell

    damage. It was reported that treatment with N-acetyl-O-methyldopamine

    (NAMDA), a metabolite of dopamine in CNS, significantly protected CA1

  • 7

    neurons in rat ischemic hippocampus and inhibited LPS-induced NO

    production in BV-2 microglia cells. In this work, NAMDA was used as a

    lead compound for NO inhibitor and the structure modification was made

    by elongation, cyclization and replacing of the acetamide group with other

    function groups. Ten compounds were synthesized and their biological

    effects were evaluated on NO production and cytotoxicity. Among all

    compounds, four compounds showed some improvements of inhibitory

    activity without increasing cytotoxicity. Compound 15 exhibited strikingly

    as the most potent NO reducing agent and more significantly potent than

    the lead compound, NAMDA.

    Recently, it was found that the bromination of 8-

    methoxyquinaldine under basic condition gave an unusual addition

    product, the 5,7-dibromo-8,8-dimethoxy-7,8-dihydroquinaldine as the

    major product via a new type reaction – electrophilic aromatic addition

    reaction: AdEAr. Upon first extension of this methodology to 1-

    methoxynaphthalene, the addition adduct was successfully obtained when

    1 equivalent of pyridine was added. This is the first reported isolation of

    an addition adduct during the electrophilic aromatic substitution of non-

    heterocyclic aromatic compounds. The importance of condition reaction

    was investigated on effect of bases and bromine sources. The reaction was

    also applied on various aromatic substrates, which yielded addition

    products as major products. Lastly, the study of mechanism indicated that

    the AdEAr reaction was occurred in anti-addition manner via cation

    intermediate. This result not only allows for the functionalization of

    aromatic compounds via the addition adducts, but also introduces the

    possibility of an alternate mechanism for electrophilic substitution

    reactions.

  • 8

    요 약 문

    생체활성물질 개발 및 아로마틱 친전자성 첨가반응에 관한

    고찰

    세로토닌 운반체나 nitric oxide synthases 에 선택적으로 강하게 결합을 하는 화합물은 선택적 세로토닌재흡수 저해제(SSRI) 역할을 하는 새로운 방사성의약품으로 혹은 NOS 저해제가 될 수 있다. 최근 발표된 선택적 세로토닌재흡수 저해제들의 다양한 구조를 바탕으로 3-(amino- and hydroxymethyl)-4-(5-iodopyridin-2-ylthio)indole 화합물들을 디자인 하였다. 이들은 세로토닌재흡수단계에서 좋은 결합을 할 것으로 예상되는 구조들을 합쳐서 디자인을 한 것이다. C3 에 치환된 4-(5-iodopyridin-2-ylthio)indoles 의 합성에 대해 기술을 하였으며, 또한 주요 중간체인 1-(5-iodopyridin-2-ylthio)-2-methyl-3-nitrobenzene (6)의 합성은 chloropyridine 7 에 thiophenol 8 로 아로마틱 친핵성치환반응에 의해서 이루어졌다. 1-(5-Iodopyridin-2-ylthio)-2-methyl-3-nitrobenzene 으로부터 개선된 Leimgruber-Batcho 인돌 합성법을 사용하여 인돌을 합성하였는데 이 과정이 목표화합물 합성 경로에서 가장 주요 반응과정이다. 불행하게도 목표화합물들과 그 유도체들은 세로토닌 운반체와 in vitro 실험에서 낮은 결합력을 가짐을 알았다. Nitric oxide (NO) 혹은 nitrogen monoxide 는 nitric oxide synthases (NOS)에 의해서 생성되는 작은 free radical 분자이다. 동물모델을 이용한 실험에서 NOS 중의 하나인 iNOS 로부터 NO 가 과생산이 될 수 있는데 이는 이스키믹상태로 인한 신경손상에 따른 생리학적 변화로 나타나며, 이는 iNOS 를 저해하므로 막을 수가 있다. NO 의 과생산을 조절하는 연구는 세포의 손상을 막을 수가 있기 때문에 매우 중요한 연구이다. 도파민의 대사물질중의 하나인 N-acetyl-O-methyldopamine (NAMDA)은 히포캠프스가 이스키믹 상태인 쥐의 CA1 neurons 의 손상을 막아주는 연구가 발표되었으며 또한 BV-2 microglia 세포에서 LPS 로 유도된 NO 의 과생산을 저해한다고 알려졌다. 이 논문에서 NAMDA 를 NO 의 생산을 저해할 수 있는 선도화합물로 사용하여, 탄소의 길이조정, 고리화 등 구조를 변화한

  • 9

    유도체를 합성하였다. 열개의 유도체를 합성하여 NO 의 생성 및 세포독성과 같은 생체활성조사를 하였다. 유도체중 네개는 세포독성의 증가없이 NO 의 생성 저해효과가 나타났으며, 그중 화합물 15 는 선도화합물 NAMDA 에 비해 가장 좋은 저해효과를 보여주고 있다. 최근 염기조건에서 8-methoxyquinaldine 을 브롬화시킬 때 치환반응대신에 첨가반응이 진행된 5,7-dibromo-8,8-dimethoxy-7,8-dihydroquinaldine 을 주 생성물로 분리할 수 있었으면 이는 새로운 형태의 반응 즉 아로마틱 친전자성 첨가반응 AdEAr 을 본 연구실에서 찾았다. 1-Methoxynaphthalene 에 이를 시도하였으며 1 당량의 피리딘을 넣어서 성공적으로 첨가반응이 진행된 생성물을 얻을 수 있었다. 이것은 헤테로원소가 없는 아로마틱 화합물에 친전자성 첨가 반응이 진행된 생성물에 대한 처음 보고이다. 이 반응에 대한 자세한 연구와 여러 가지 유도체에 대한 일반화 시키는 것에 대한 연구를 진행하였다. 그리고 반응메카니즘에 대한 연구로부터 anti-addition 에 의해 반응이 진행됨을 알았다. 첨가생성물로부터 다양한 유도체를 합성할 수가 있으며, 또한 친전자성 치환반응의 새로운 메커니즘을 제시할 가능성을 언급하였다.

  • 1

    PART 1

    The Syntheses of 3-Substituted 4-(Pyridin-2-ylthio)indoles

    via Leimgruber-Batcho Indole Synthesis

  • 2

    Introduction

    Serotonin or 5-hydroxytryptamine is the baby-boomer of

    neurotransmitters. Serotonin has been associated with, among other things,

    anxiety, depression, schizophrenia, drug abuse, sleep, dreaming,

    hallucinogenic activity, headache, cardiovascular disorders, appetite

    control, and is now dabbling in acupuncture and transcendental

    meditation.1 A review of the recent patent literature provides an indication

    of some of the newer claim being made for novel serotonergic agents.

    Tens of thousands of papers have been published on serotonin; much is

    known –but an incredible amount remains to be learned.

    NH

    NH2

    HO

    Serotonin (5-HT)

    Serotonin biosynthesis, catabolism and function as targets for drug

    manipulation2

    5-HT is biosynthesized from its dietary precursor L-tryptophan

    (Scheme 1). Serotonergic neurons contain tryptophan hydroxylase (L-

    trytophan-5-monooxygenase) that converts tryptophan to 5-

    hydroxytryptophan (5-HTP) in what is the rate-limiting step in 5-HT

    biosynthesis, and aromatic L-amino acid decarboxylase (previously called

    5-HTP decarboxylase) that decarboxylates 5-HTP to 5-HT. This latter

    enzyme is also responsible for the conversion of L-DOPA to dopamine.

  • 3

    The major route of metabolism for 5-HT is oxidative deamination by

    monoamine oxidase (MAO-A) to the unstable 5-hydroxylindole-3-

    acetaldehyde which is either reduced to 5-hydroxytryptophol (~15%) or

    oxidized to 5-hydroxyindole-3-acetic acid (~85%). In the pineal gland, 5-

    HT is acetylated by 5-HT N-acetyltransferase to N-acetylserotonin, which

    undergoes O-methylation by 5-hydroxyindole-O-methyltransferase to

    melatonin.

    NH

    NH2

    HO

    MAO

    NH

    HN

    HO

    Ac

    NH

    NH2HOOC

    HO

    NH

    OHC

    HO

    NH

    OH

    HO

    NH

    HOOC

    HO

    NH

    HN

    H3CO

    Ac

    NH

    NH2HOOC

    +

    Tryptophan 5-Hydroxytryptophan(5-HTP)

    Serotonin(5-HT)

    Melatonin N-acetyl-5-HT

    5-Hydroxyindole-3-acetaldehyde

    5-Hydroxyindole-3-acetic acid

    5-Hydroxytryptophol

    Tryptophan

    Hydroxylase

    Aromatic

    amino aciddecarboxylase

    5-HT N-acetyl-transferase

    5-Hydroxyindole-

    O-methyltransferase

    Scheme 1. Biosynthesis and catabolism of serotonin.

  • 4

    Each of the steps in 5-HT biosynthesis, metabolism, and function

    is a theoretical target for drug manipulation. Tryptophan depletion, by

    reducing or eliminating dietary tryptophan, can result in decreased 5-HT

    biosynthesis. Conversely, tryptophan “loading,” by increasing dietary

    tryptophan, can result in the overproduction of 5-HT. This latter effect can

    also occur in non-serotonergic neurons, such as dopaminergic neurons,

    because of the non-selective nature of aromatic amino acid decarboxylase.

    Inhibitors of trytophan hydroxylase such as para-chlorophenylalanine are

    used as pharmacologic tools and are not used therapeutically.

    Serotonin reuptake transporter

    Transporter proteins are specific to their respective transmitter. In

    the case of serotonin, a transporter protein found in the plasma membrane

    of serotonergic neurons is responsible for re-uptake of the transmitter. The

    transporter protein acts as a carrier of serotonin molecules across the

    membrane. Unlike channels, which stay open or closed, transporters

    undergo conformational changes (changes in their three dimensional

    shape) and move one molecule in each cycle.

    The serotonin transporter (SERT) is similar to other biogenic

    amine transporters (e.g. norepinephrine and dopamine transporters) and is

    part of a family of sodium (Na+) and chloride (Cl-) dependent transporters.

    SERTs, with molecular weights of 60-80 kDA, have twelve

    transmembrane domains (TM) with a large extracellular loop between

    TMs 3 and 4. Both the N- and C-termini are located within the cytoplasm

    (Figure 1). The large extracellular loop and the intracellular parts of the N-

  • 5

    and C-termini do not appear to be the significant sites that determine

    interactions with 5-HT or transporter inhibitors. Rather, the areas

    important for selective 5-HT affinity appear to be localized within TMs 1-

    3 and TMs 8-12. It is believed that SERTs have a common binding site for

    5-HT and many of its inhibitors. SERT activity, like serotonin, is seen

    most often in the raphe nuclear complex. SERTs have also been seen in

    the amygdale, thalamus, hypothalamus, substantial nigra, and locus

    coeruleus. In addition to being found on neurons, SERTs are seen in the

    placenta, lungs, and blood platelets. Blood platelets utilize SERTs to

    obtain serotonin from the environment because they cannot synthesize it

    themselves. In the placenta, SERTs may protect heavily vascularized

    embryonic tissue from constricting too early due to maternal serotonin.

    Figure 1. Structure of serotonin reuptake transporter

    Serotonin reuptake transporters (SERTs) are dependent on

    extracellular Na+ and extracellular Cl-. Unlike Na+, Cl- can be at least

    partly substituted for by NO2-, Br-, and other anions. Intracellular

  • 6

    potassium (K+) is also used in the process but can be replaced by other

    ions, most notably hydrogen (H+). The driving force for the energetically

    unfavorable transport of serotonin is the Na+ influx down its concentration

    gradient. The Na+/K+ pump (Na+/K+ ATPase) maintains the extracellular

    Na+ concentration as well as the intracellular K+ concentration. Na+/K+

    ATPase pumps three Na+ ions our for each two K+ ions pumped into the

    cell. The electrical potential produced, in addition to creating the Na+

    concentration used by the transporter protein, also leads to the loss of Cl-

    ions from the cell, which is also used in transport.

    According to the present model of SERT function, the first step

    occurs when Na+ binds to the carrier protein. Serotonin, in its protonated

    form (5-HT+), then binds to the transporter followed by Cl-. Chloride ions

    are not required for 5-HT+ binding to occur but are necessary for net

    transport to take place. The initial complex of serotonin, Na+, and Cl-

    creates a conformational change in the transporter protein. The protein,

    which began by facing the outside of the neuron, moves to an inward

    position where the neurotransmitter and ions are released into the

    cytoplasm of the neuron. Intracellular K+ then binds to the SERT to

    promote reorientation of the carrier for another transport cycle. The

    unoccupied binding site becomes, once again, exposed to the outside of

    the cell and the K+ is released outside the cell.

    Serotonin reuptake inhibitor

    The reuptake process is susceptible to drug manipulation. By

    blocking the action of serotonin reuptake transporter, the amount of

    serotonin in the synaptic cleft increases. Selective serotonin reuptake

  • 7

    inhibitors (SSRIs) act primarily at the 5-HT transporter protein and have

    limited, if any, reaction with other neurotransmitter systems. SSRIs bind

    to the transporter protein directly and block the reuptake process.

    Consequently, more serotonin remains in the cleft where it is free to travel

    further to more distant receptors as well as continue to react with nearby

    receptors. Like the binding of substrates, antagonist binding to SERTs is

    also dependent on extracellular Na+ although ion dependency is different

    for each SERT antagonist. It is unclear whether SSRIs bind to the same

    SERT domain as serotonin or operate through more indirect mechanisms.

    Recent evidence suggests that binding of SSRIs to SERTs occurs at the

    same site as 5-HT binding, but it has not been determined conclusively.

    In the late-1980s, the serotonin-selective reuptake inhibitor (SSRI)

    fluoxetine became the mainstay of treatment for clinical depression-

    replacing the more toxic tricyclic antidepressants (TCAs).3 SSRIs have a

    more favorable adverse reaction profile in comparison to the TCAs and

    are much easier to tolerate. SSRIs are the focus of extensive research into

    finding beneficial pharmacological therapies. There is evidence that

    serotonergic pathways are the most closely related systems to mood

    disorders, especially depression, and thus SSRIs may lead to significant

    therapy.4 Clinical depression is one of the most common psychiatric

    disorders, with an incidence of about 4% and a life-time prevalence of 15-

    20%. Despite significant research, the neurobiological dysfunctions of

    major depression remain elusive. Findings implicate multiple

    abnormalities in serotonergic pathways in the cause of depression.

    Findings include 1) low concentrations of the major serotonin metabolite;

    2) a low density of brain and platelet serotonin transporters in depressed

    individuals; 3) a high density of brain and platelet serotonin binding sites;

  • 8

    and 4) a low concentration of tryptophan, which is used in serotonin

    synthesis. Of these, the low level of SERTs in depressed patients has

    received the most attention. Still, while the precise cause of depression

    eludes neuroscientists, SSRIs have been shown to alleviate the mood

    disorder and are a common therapy for depression. There are many SSRIs

    either in the market or in development.5 SSRIs currently available include

    citalopram, fluoxetine (Prozac), fluvoxamine, paroxetine (Paxil), and

    sertraline (Zoloft). In addition, while not as selective as the above-

    mentioned, drugs of abuse such as cocaine, fenfluramine, and (3,4-

    methylenedioxy) methamphetamine (MDMA or ecstasy) are inhibitors of

    serotonin uptake.

    SSRI imaging

    Imaging of SERT in humans would provide a useful tool to

    understand how alterations of this system are related to depressive illness

    and other psychiatric disorders; therefore, it potentially can benefit

    millions of patients who are being treated with SSRIs. The first successful

    radioligand was [11C](+)McN5652 (1) for positron emission tomography

    (PET) imaging (Chart 1).6,7 It showed excellent inhibition of 5-HT

    reuptake in rat brain synaptosomes (Ki = 0.40 nM for inhibition of SERT)

    and moderate selectivity toward other monoamine transporters (DAT,

    dopamine, and NET, norepinephrine transporters; Ki 23.5 and 1.82 nM,

    respectively).8 Specific binding of [11C](+)McN5652 correlates well with

    the known density of SERT sites in the human brain.6,9,10 Recent reports,

    using [11C](+)McN5652 for imaging SERT as an indicator of serotonin

    neurons, have suggested that MDMA (methylenedioxymethamphetamine,

    “ecstasy”) may cause an irreversible decrease of SERT binding sites.11

  • 9

    (Despite its successful demonstration in imaging SERTs in humans, it has

    been reported previously that [11C](+)McN5652 has several limitations.10)

    The uptake in the specific binding area is slow requiring at least 120 min

    of data acquisition. The nonspecific binding is relatively high, which

    precludes the measurements of lower SERT density regions. The plasma

    free fraction is very low (

  • 10

    SERT over NET and DAT (Ki ) 699 and 840 nM, for NET and DAT,

    respectively). A preliminary imaging study of [123I]- ADAM in the brain

    of a baboon by SPECT at 180-240 min post iv injection indicated a

    specific uptake in the midbrain region rich in SERT. It is apparent that

    [123I]- ADAM showed a significant improvement over [123I]- IDAM as a

    SPECT imaging agent for SERT in the brain.17,20 Initial imaging studies in

    humans suggest that the agent clearly localized in the region of

    hypothalamus region of the brain where the concentration of SERT is the

    highest.

    N

    O2NI

    NNH

    I

    S

    N

    NH2

    I

    S

    N

    OH

    N S

    I

    NH

    R

    N

    SCH3

    1, (+)McN5652 2, 5-iodoquipazine 3, ADAM

    4, IDAM 5 Chart 1

    Although these radioligands all serve as potent selective ligands of

    SERT, they bear significant structural differences from one another. In

    light of their combined success, and in an effort to optimize structural

    contributions to SERT binding,21 we have designed a new type of

  • 11

    radioligand, 4-(pyridin-2-ylthio)indole 5 (Chart 1), as a combination of the

    various structural moieties characteristic of these potent inhibitors by

    computer designing from 3D structure of SERT binding site. The designed

    target compound 5 is synthetically challenging due to the lack of available

    methodology for the formation of heterodiaryl sulfides, specifically 4-

    thioindoles. After attempt of various synthetic routes, the Leimgruber-

    Batcho indole synthesis22 was finally selected as the key step in converting

    precursor 6 to indole 5 because of its mild reaction conditions and

    regioselectivity for 4-substituted indoles (see Figure 2 for retrosynthetic

    analysis). The synthesis of key intermediate 6 occurs by nucleophilic

    aromatic substitution of chloropyridine 7 with thiophenol 8. The synthesis

    of 2-chloro-5-iodopyridine (7) by iodination23 and Sandmeyer reaction24

    was reported whereas thiophenol 8 is a new compound. Although it is

    possible to directly convert aniline derivative to thiolphenol by

    Sandmeyer-type reaction, violently explosive diazo sulfides and related

    compounds may be formed, and another less hazardous method for the

    preparation of desired compound should be used, if possible.25 Therefore,

    commercially available 2-methyl-3-nitroaniline would be diazotized to

    phenol derivative and then finally converted to thiolphenol 8 by Newman

    and Karnes’ method.26 Herein, we wish to report the synthesis of 4-

    (pyridin-2-ylthio)indole analogues in detail.

  • 12

    N S

    I

    NH

    R

    5

    N S

    I

    NO2

    N Cl

    I

    NO2

    SH

    +

    6

    7 8

    N NH2NO2

    NH2

    Figure 2. The retrosynthesis of 3-substituted 4-(5-iodopyridin-2-ylthio)indoles.

  • 13

    Results and Discussion

    2-Chloro-5-iodopyridine (7) (Scheme 2) was prepared as previously

    reported by conversion of commercially available 2-aminopyridine (9) to

    2-amino-5-iodopyridine (1023) with periodic acid and iodine, followed by

    halogenation via the diazonium salt to give 724 in 41% yield.

    Preparation of 2-methyl-3-nitrothiophenol was successfully achieved

    by the method outlined in Scheme 3. Aniline 11 was first diazotized to

    phenol 12 in excellent yield. Treatment of 12 with dimethylthiocarbamoyl

    chloride in the presence of KOH provided thionocarbamate 13, which was

    then thermally converted to thiocarbamate 14 in a sealed tube by Newman

    and Karnes’ condition.26 Hydrolysis of 14 afforded thiol 8 with a trace

    amount of disulfide 15 as by-product.

    N Cl

    I

    N NH2 N NH2

    Ia b

    9 10 7 Scheme 2. (a) I2, H5IO6, AcOH, H2SO4, 80 °C, 1 h, 65%; (b) NaNO2, HCl, CuCl,

    0 °C-rt, overnight, 41%.

  • 14

    NO2

    S 2

    NH2

    NO2

    S

    NO2

    O

    Me2N

    OH

    NO2

    SH

    NO2

    O

    NO2

    S

    Me2N

    11 12 13

    14 8 15

    a b

    c d+

    Scheme 3. (a) NaNO2, H2SO4, H2O, 0-5 °C, 10 min, 120 °C, 5 min, 94%; (b)

    N,N-dimethylthiocarbamoyl chloride, KOH, THF, H2O, 0 °C-rt, 30 min, 76%; (c)

    220 °C in a sealed tube, 3 h, 80%; (d) KOH, H2O, MeOH, 80 °C, 1 h, (8, 95%)

    (15, trace)

    Chloropyridine 7 and thiol 8 were subsequently used as starting

    materials in the preparation of 4-(pyridin-2-ylthio)indole analogues as

    shown in Scheme 4. The iodo moiety of 7 precluded the use of metal

    catalysts for the coupling reaction between 7 and 8, but the reaction

    proceeded quite nicely when executed in a sealed tube under N2 to give

    sulfide 6 in the absence of catalyst. Unfortunately, under these conditions

    we also obtained a by-product disulfide 15, which proved difficult to

    remove from the major product 6. Interestingly, the addition of

    triphenylphosphine to the reaction mixture not only prevented disulfide

    formation, but also permitted the use of the thiophenol/disulfide mixture

    as a suitable starting material under these conditions.

  • 15

    S

    NO2

    N

    I

    6

    a

    SN

    I

    NH

    CHO

    19

    b

    SN

    I

    NH

    NH2

    SN

    I

    NH

    OH

    20

    21

    d

    e

    SH

    NO28 (with trace amount 15)

    c

    SN

    I

    NH

    +

    16

    SN

    I

    NH

    O

    17

    +

    18

    NH2

    S N

    I

    Scheme 4. (a) 7, PPh3, Et3N, DMF, 110 °C, 20 h, 71%; (b) (i)

    dimethylformamide dimethyl acetal (DMF-DMA), pyrrolidine, DMF, 110 °C, 2

    h, (ii) Fe, AcOH, 100 °C, 3h, (16, 28%, 17, 11%, 18, 13%); (c) POCl3, DMF,

    40oC, 1 h, 40%; (d) NaCNBH3, NH4OAc, MeOH, rt, 3 days, 20%; (e) NaBH4,

    EtOH, rt, 4 h, 52%.

  • 16

    The modified Leimgruber-Batcho indole synthesis (Scheme 4) was

    attempted under a variety of conditions. The first effort, which utilized

    acetic acid as solvent in the reduction step, afforded the desired indole 16

    as well as acetyl derivative 17 and thiophene 18 as by-products. The

    structure of 18 was confirmed by spectroscopic data, including 2D-

    HETCOR NMR, which was assigned structurally in Figure 3, and mass

    spectrometry. (A plausible mechanism for the formation of thiophene is

    shown in Scheme 5). A subsequent attempt using HCl/EtOH as solvent

    gave only trace amounts of target product 16 and some unidentified

    residue. After a considerable number of trials, an eventual solvent mixture

    of AcOH/EtOH (1:1) proved optimal, affording product 16 and

    dramatically decreasing the amount of by-product formed. Formylation of

    16 via the Vilsmeier reaction yielded aldehyde 19, which subsequently

    underwent either reductive-amination or reduction to give 3-

    aminomethylindole 20 and 3-hydroxymethylindole 21 respectively.

    8.86

    7.46 8.05

    7.28

    7.186.64

    5.11NH2

    S N

    I

    NH2

    S N

    I

    7.40 127.0

    145.2

    154.3

    126.0

    126.0

    112.5

    110.0

    Figure 3 Assignments of 1H and 13C NMR based on 2D-HETCOR:

  • 17

    SN

    I

    NO2

    -H+ SN

    I

    NO2

    N+MeO

    SN

    I

    NO2

    NMe2

    S

    NO2

    NMe2

    N

    I

    H

    S

    NO2

    N

    H NMe2

    I

    NO2

    S N

    I

    NH2

    S N

    I

    S

    NO2

    NMe2

    NI

    15

    -HNMe2

    reduction

    18 Scheme 5. Plausible Mechanism of the Formation of Thiophene 18.

  • 18

    Conclusions

    In conclusion, a series of 4-(pyridin-2-ylthio)indole derivatives have

    been successfully synthesized for serotonin transporter imaging agent

    evaluation. The conversion of 2-methyl-3-nitrophenol (12) to 2-methyl-2-

    nitrothiophenol (8) was took place via thionocarbamate intermediate,

    which was subjected to thermal rearrangement. Addition of

    triphenylphosphine in the coupling reaction of 7 and 8 effectively

    prevented disulfide formation. A modified Leimgruber-Batcho indole

    synthesis was accomplished from the key intermediate 1-(5-iodopyridin-2-

    ylthio)-2-methyl-3-nitrobenzene (6). The heterodiaryl sulfide chemistry

    utilized in this synthesis could be useful for the preparation of other novel

    bioactive compounds.

  • 19

    Experimental Section

    2-Amino-5-iodopyridine (10). A mixture of 2-aminopyridine (2.06 g,

    21.9 mmol), acetic acid (14 mL), water (3 mL), sulfuric acid (0.42 mL),

    and H5IO6 (1.05 g, 4.6 mmol) was allowed to stir at 80 °C for 15 min.

    Iodine crystals (2.28 g, 9.0 mmol) were added in portions. After it was

    stirred for 1 h, the reaction mixture was poured into saturated sodium

    thiosulfate solution and extracted with ethyl acetate. The organic layer was

    separated, dried (Na2SO4), and evaporated to give 10 (3.13 g, 65%) as an

    orange solid: 1H NMR (200 MHz, CDCl3) δ 4.85 (br, 2H), 6.36 (dd, J =

    8.8, 0.8 Hz, 1H), 7.64 (dd, J = 8.8, 2.2 Hz, 1H), 8.18 (d, J = 2.0 Hz, 1H).

    CAS No. 20511-12-0.

    2-Chloro-5-iodopyridine (7). A mixture of aminopyridine 10 (1.05 g,

    4.74 mmol) and concentrated HCl (10 mL) was stirred at 0 °C for 10 min.

    Sodium nitrite (1.38 g, 20.0 mmol) was slowly added, then followed by

    CuCl (0.50 g, 5.1 mmol) with stirring continued overnight. The mixture

    was poured into 1:1 NH4OH:H2O, extracted with ethyl acetate, dried

    (Na2SO4), and concentrated. The crude residue was purified by flash

    column chromatography on silica gel using pure dichloromethane as an

    eluant to yield 7 (0.47 g, 41%) as a colorless solid: 1H NMR (200 MHz,

    DMSO-d6) δ 7.36 (d, J = 8.4 Hz, 1H), 8.18 (dd, J = 8.2, 2.2 Hz, 1H), 8.64

    (d, J = 1.6 Hz, 1H); 13C NMR (50 MHz, DMSO-d6) δ 93.1, 126.8, 148.0,

    150.2, 155.9; CAS No. 69045-79-0.

    2-Methyl-3-nitrophenol (12). To a mixture of 11 (3.8 g, 25.0 mmol),

    concentrated sulfuric acid (5.5 mL) and water (7.5 mL), 20 g of ice was

    added and the solution was cooled to 0-5 °C. A solution of sodium nitrite

  • 20

    (1.8 g, 26 mmol) in 1.5 mL of water was added. After stirred for 10 min,

    the mixture was allowed to stand at 0-5 °C for 5 min. To a boiling solution

    of concentrated sulfuric acid (16.5 mL) and water (15 mL), the diazotized

    solution was slowly added. After adding, the mixture was boiled for 5 min

    and then poured to a beaker containing ice-water. The precipitate was

    collected by suction filtration, washed with cold water and dried. The solid

    was purified by flash column chromatography (EtOAc:hexane, 1:9) to

    yield 3.63 g (94%) of 12 as a yellow solid: 1H NMR (200 MHz, DMSO-

    d6) δ 2.23 (s, 3H), 7.12 (dd, J = 8.2, 1.8 Hz, 1H), 7.19 (dd, J = 8.2, 7.8 Hz,

    1H), 7.44 (dd, J = 7.8, 1.8 Hz, 1H); 13C NMR (50 MHz, DMSO-d6) δ 11.8,

    114.7, 119.3, 127.4, 151.5, 157.2; CAS No. 5460-31-1

    2-Methyl-3-nitrophenyl N,N-Dimethylthionocarbamate (13). To a

    powder of 12 (3.06 g, 20.0 mmol) was added a solution of potassium

    hydroxide (1.12 g, 20.0 mmol) in 15 mL of H2O at rt. The mixture was

    cooled below 5 °C in ice-water bath. A solution of N,N-

    dimethylthiocarbamyl chloride (0.185 g, 1.5 mmol) in 5 mL of dry THF

    was added with cooling. After the addition, the reaction mixture was

    allowed to stir at rt for 30 min. The mixture made alkaline with 10%

    potassium hydroxide and extracted with dichloromethane. The organic

    layers are combined, washed with brine, and dried over. The residue was

    purified by flash column chromatography (CH2Cl2:hexane, 8:2) to give 13

    (3.67 g, 76%) as an yellow solid: 1H NMR (200 MHz, CDCl3) δ 2.36 (s,

    3H), 3.40 (s, 3H), 3.48 (s, 3H), 7.26 (dd, J = 8.0, 1.4 Hz, 1H), 7.36 (t, J =

    8.0 Hz, 1H), 7.83 (dd, J = 8.0, 1.4 Hz, 1H); 13C NMR (50 MHz, CDCl3) δ

    13.0, 39.0, 43.7, 122.3, 126.6, 127.5, 128.5, 150.7, 153.5, 186.4. MS (EI)

    240 (M+), 225, 223, 194, 179, 151, 121, 88, 72 (100), 63, 51. HRMS (EI)

    Calc. for C10H12N2O3S (M+) 240.0569. Found 240.0564.

  • 21

    2-Methyl-3-nitrophenyl N,N-Dimethylthiocarbamate (14). A powder of

    13 (1.57 g, 6.5 mmol) was added into a sealed tube and purged with N2.

    The tube was capped and heated at 215-220 °C for 3 h. The reaction was

    cooled to rt. The residue was purified by flash column chromatography

    (CH2Cl2) to provide 14 (1.24 g, 80%) as an orange solid: 1H NMR (200

    MHz, CDCl3) δ 2.59 (s, 3H), 3.03 (br, 3H), 3.14 (br, 3H), 7.33 (dd, J = 7.8,

    7.6 Hz, 1H), 7.75 (dd, J = 7.6, 1.4 Hz, 1H), 7.85 (dd, J = 8.0, 1.4 Hz, 1H); 13C NMR (50 MHz, CDCl3) δ 17.7, 37.3, 125.9, 126.7, 132.5, 137.7, 141.7,

    151.5, 165.2; MS (EI) 240 (M+), 210, 168, 149, 121, 110, 72 (100), 56.

    HRMS (EI) Calc. for C10H12N2O3S (M+) 240.0569. Found 240.0570.

    2-Methyl-3-nitrothiophenol (8). A solution of 14 (1.24 g, 5.2 mmol) and

    KOH (0.60 g, 10.7 mmol) in H2O (2 mL) and MeOH (10 mL) was heated

    at 80 °C for 1 h under N2 atmosphere. The reaction mixture was cooled

    and poured to 5 g of ice. The solution was washed with CH2Cl2 (20 mL x

    2). The organic layer was discarded. The aqueous layer was acidified by 4

    M HCl solution and extracted with CH2Cl2 (20 mL x 2). The organic

    layers were combined, dried (Na2SO4) to give a mixture of 8 (0.84 g,

    95%): 1H NMR (200 MHz, CDCl3) δ 2.44 (s, 3H), 3.58 (s, 1H), 7.17 (t, J

    = 8.0 Hz, 1H), 7.49 (d, J = 8.0 Hz, 1H), 7.55 (dd, J = 8.0, 0.8 Hz, 1H); 13C

    NMR (50 MHz, CDCl3) δ 17.2, 121.6, 126.9, 130.0, 133.7, 135.8, 151.6;

    MS (EI) 169 (M+), 152 (100), 124, 121, 110, 97, 77, 63, 45, 39; HRMS

    (EI) Calc. for C7H7NO2S (M+) 169.0198. Found 169.0195. Di(2-methyl-

    3-nitrophenyl) Disulfide (15). (trace amount) 1H NMR (200 MHz,

    CDCl3) δ 2.60 (s, 6H), 7.29 (dd, J = 8.2, 8.0 Hz, 1H), 7.69 (d, J = 8.0 Hz,

    1H), 7.71 (dd, J = 8.2, 8.0 Hz, 1H); 13C NMR (50 MHz, CDCl3) δ 16.3,

    123.6, 127.3, 131.7, 132.3, 138.4, 151.5; MS (EI) 336 (M+, 100), 320, 306,

  • 22

    259, 241, 168, 129, 121, 110, 77, 57. HRMS (EI) Calc. for C14H12O4N2S2

    (M+) 336.0239. Found 336.0242.

    1-(5-Iodopyridin-2-ylthio)-2-methyl-3-nitrobenzene (6). A mixture of 8

    and 15 (0.84 g, 4.97 mmol), 7 (1.19 g, 4.97 mmol), PPh3 (0.13 g, 0.5

    mmol) and Et3N (0.72 mL) in DMF (5 mL) was added into a sealed tube

    and purged with N2. The reaction was heated at 110 °C for 20 h. The

    mixture was cooled, added CH2Cl2 (20 mL) and washed with H2O and

    brine. The organic layer was dried (Na2SO4) and evaporated. The residue

    was purified by flash column chromatography (EtOAc:hexane, 8:2) to

    give 6 (1.32 g, 71%) of as a yellow solid: 1H NMR (200 MHz, CDCl3) δ

    2.56 (s, 3H), 6.77 (d, J = 8.4 Hz, 1H), 7.37 (dd, J = 8.0, 7.8 Hz, 1H), 7.76-

    7.82 (m, 2H), 7.87 (d, J = 8.0 Hz, 1H), 8.59 (d, J = 2.2 Hz, 1H); 13C NMR

    (50 MHz, CDCl3) δ 17.5, 89.1, 123.5, 125.8, 127.4, 133.8, 137.1, 140.5,

    145.2, 151.9, 156.1, 158.3; MS (EI) 372 (M+), 357, 353, 327, 311, 227,

    197, 154, 121, 89, 77, 63, 51, 39. HRMS (EI) Calc. for C12H9N2O2SI (M+)

    371.9430. Found 371.9426.

    4-(5-Iodopyridin-2-ylthio)indole (16). A mixture of 6 (1.72 g, 4.6 mmol),

    dimethylformamide dimethyl acetal (1.4 mL, 10 mmol) and pyrrolidine

    (0.4 mL, 5 mmol) in 8 mL of DMF was allowed to heat at 110 °C for 2 h

    under N2 atmosphere. The reaction was cooled, added ether (20 mL) and

    washed with H2O (20 mL x 2). The organic layer was dried over, and

    evaporated. The red residue was dissolved in the mixture of AcOH (15

    mL) and EtOH (15 mL) and added iron powder (2 g). The suspension was

    heated at 100 °C for 3 h. The reaction was cooled, filtered and washed by

    water. The filtrate was basified by 1 M NaOH solution and extracted with

    ether, washed with H2O and brine. The extract was dried over and was

  • 23

    purified by flash column chromatography (ethyl acetate:hexane, 1:4) to

    provide 16 (0.62 g, 36%) as a off-white solid: 1H NMR (200 MHz,

    DMSO-d6) δ 6.27-6.29 (m, 1H), 6.43 (d, J = 8.4 Hz, 1H), 7.19 (dd, J = 7.6,

    7.4 Hz, 1H), 7.32 (dd, J = 7.2, 1.0 Hz, 1H), 7.44 (dd, J = 3.0, 2.4 Hz, 1H),

    7.59 (dd, J = 8.0, 1.0 Hz, 1H), 7.83 (dd, J = 8.4, 2.2 Hz, 1H), 8.60 (d, J =

    2.2 Hz, 1H) 11.49 (br, 1H); 13C NMR (50 MHz, DMSO-d6) δ 88.7, 100.4,

    113.9, 119.0, 121.8, 122.3, 126.7, 127.0, 130.5, 136.3, 144.8, 154.7,

    160.2; MS (EI) 352 (M+), 224 (100), 207, 147, 104, 77, 73, 50. HRMS

    (EI) Calc. for C13H9N2SI (M+) 351.9531. Found 351.9529.

    3-Acetyl-4-(5-iodopyridin-2-ylthio)indole (17) and 3-(4-Iodopyridin-

    2yl)-4-aminothiophene (18). Same procedure used as 16, pure AcOH (30

    mL) was used instead of the mixture of AcOH and EtOH in the reduction

    step to provide 16 (0.48 g, 28%), 17, and 18. 17 (0.20 g, 11%) as a white

    solid: 1H NMR (200 MHz, CDCl3) δ 2.05 (s, 3H), 7.40 (dd, J = 8.2, 7.6 Hz,

    1H), 7.52 (d, J = 8.4 Hz, 1H), 7.63 (dd, J = 8.0, 1.0 Hz, 1H), 7.62 (s, 1H),

    8.12 (dd, J = 8.4, 2.2 Hz, 1H), 8.27 (d(br), J = 7.4 Hz, 1H), 8.88 (d, J = 2.2

    Hz, 1H) 11.45 (br, 1H); 13C NMR (50 MHz, CDCl3) δ 25.0, 91.4, 118.5,

    118.9, 126.0, 126.6, 127.2, 130.2, 135.2, 135.7, 142.6, 146.6, 153.6, 155.0,

    168.4; MS (EI) 394 (100, M+), 379, 352, 281, 224, 209, 197, 121, 73, 50;

    HRMS (EI) Calc. for C15H11N2OSI (M+) 393.9637. Found 393.9636. 18

    (0.23 g, 13%) as a white solid: 1H NMR (400 MHz, CDCl3) δ 5.11 (br,

    2H), 6.64 (dd, J = 7.6, 1.2 Hz, 1H), 7.18 (dd, J = 8.0, 7.6 Hz, 1H), 7.28

    (dd, J = 8.0, 1.2 Hz, 1H), 7.40 (s, 1H), 7.46 (dd, J = 8.2, 0.8 Hz, 1H), 8.05

    (dd, J = 8.2, 2.4 Hz, 1H), 8.86 (dd, J = 2.4, 0.8 Hz, 1H); 13C NMR (100

    MHz, CDCl3) δ 91.3, 111.0, 112.5, 124.0, 126.0, 127.0 136.5, 142.9,

    144.2, 145.3, 154.3, 155.2; MS (EI) 352 (M+, 100), 336, 281, 225, 224,

  • 24

    198, 121, 113, 99, 77, 57. HRMS (EI) Calc. for C13H9N2SI (M+) 351.9531.

    Found 351.9532.

    3-Formyl-4-(5-iodopyridin-2-ylthio)indole (19). Phosphorus oxychloride

    (0.05 mL, 0.27 mmol) was added dropwise with stirring to DMF (0.5 mL)

    at 0-5 °C. A solution of 16 (0.10 g, 0.28 mmol) in DMF (1 mL) was then

    added dropwise. After addition, the mixture was stirred at 40 °C for 1 h.

    The mixture was poured to 1 g of crushed ice and then made alkaline by 1

    M NaOH solution. The resulting suspension was heated to the boiling

    point and cooled to rt. The precipitate was filtered, washed by water, air-

    dried. The solid was purified by flash column chromatography

    (CH2Cl2:hexane, 6:4) to give 19 (42 mg, 40%) as a off-white solid: 1H

    NMR (400 MHz, DMSO-d6) δ 6.56 (dd, J = 8.4, 0.8 Hz, 1H), 7.33 (dd, J =

    8.0, 7.6 Hz, 1H), 7.53 (dd, J = 7.6, 0.8 Hz, 1H), 7.72 (dd, J = 8.0, 0.8 Hz,

    1H), 7.88 (dd, J = 8.4, 2.0 Hz, 1H), 8.25 (d, J = 3.2 Hz, 1H), 8.59 (dd, J =

    2.0, 0.8 Hz, 1H) 10.32 (s, 1H); 13C NMR (100 MHz, DMSO-d6) δ 89.9,

    116.0, 118.9, 120.1, 123.0, 124.0, 128.2, 131.7, 134.2, 138.4, 145.8, 155.8,

    160.0, 186.0; MS (EI) 380 (M+), 351, 347 (100), 320, 224, 176, 148, 129,

    104, 88, 72, 57. HRMS (EI) Calc. for C14H9N2OSI (M+) 379.9480. Found

    379.9477.

    3-(Aminomethyl)-4-(5-iodopyridin-2-ylthio)indole (20). To a solution of

    19 (0.10 g, 0.26 mmol), ammonium acetate (0.20 g, 2.6 mmol) in 4 mL of

    MeOH was added a solution of 1.0 M NaCNBH3 in THF (0.26 mL, 0.26

    mmol) with stirring. The reaction was stirred at rt for 3 days. The reaction

    was acidified by 2 M HCl solution until pH

  • 25

    (CH2Cl2:MeOH, 10:1) to give 20 (20 mg, 20%) of as a white solid: 1H

    NMR (400 MHz, DMSO-d6) δ 4.27 (br, 2H), 6.37 (d, J = 8.4 Hz, 1H),

    7.22 (dd, J = 8.0, 7.6 Hz, 1H), 7.28 (d, J = 7.2 Hz, 1H), 7.64 (d, J = 8.0 Hz,

    1H), 7.74 (d, J = 2.8 Hz, 1H), 7.81 (dd, J = 7.6, 2.4 Hz, 1H), 8.43 (d, J =

    2.4 Hz, 1H), 9.22 (br, 1H), 12.04 (br, 1H); 13C NMR (100 MHz, DMSO-

    d6) δ 42.0, 90.2, 105.8, 115.3, 119.6, 122.9, 123.1, 128.3, 130.2, 130.7,

    137.8, 145.7,155.5, 161.0 MS (FAB) 382 (M+), 380, 365 (100), 332, 239,

    207, 115. HRMS (FAB) Calc. for C14H13N3SI (M+H+) 381.9875. Found

    3819871.

    3-(Hydroxymethyl)-4-(5-iodopyridin-2-ylthio)indole (21). A suspension

    of 19 (38 mg, 0.10 mmol), NaBH4 (4.0 mg, 0.1 mmol) in 0.6 mL of 95%

    EtOH was stirred at rt for 4 h. The reaction mixture was added with water

    and extracted with ethyl acetate. The organic layers were combined, then

    washed by water and brine, and dried (Na2SO4). The crude was purified by

    flash column chromatography (EtOAc:hexane, 3:7)to provide 21 (20 mg,

    52%) as an off-white-off solid: 1H NMR (400, DMSO-d6) δ 4.63-4.67 (m,

    3H), 6.35 (dd, J = 8.8, 0.4 Hz, 1H), 7.17 (dd, J = 8.0, 7.2 Hz, 1H), 7.25

    (dd, J = 7.2, 1.2 Hz, 1H), 7.34 (dd, J = 1.6, 1.2 Hz, 1H), 7.55 (dd, J = 8.0,

    2.0 Hz, 1H), 7.85 (dd, J = 8.8, 2.4 Hz, 1H), 8.59 (dd, J = 2.2, 0.4 Hz, 1H),

    11.31 (d, J = 2.0 Hz, 1H); 13C NMR (100 MHz, DMSO-d6) δ 57.2, 89.1,

    114.8, 117.6, 119.4, 122.4, 122.5, 126.0, 128.1, 129.0, 138.3, 145.6, 155.3,

    162.6; MS (FAB) 383 (M+), 365 (100), 332, 239, 160, 117. HRMS (FAB)

    Calc. for C14H12N2OSI (M+H+) 382.9715. Found 382.9713..

  • 26

    References

    1. a) Williams, D. A.; Lemke, T. L. Foye’s Principles of Medicinal

    Chemistry 5th ed.; Lippincott Williams&Wilkins: Baltimore, 2002.

    b) Rapport, M. R. Perspect. Bio. Med. 1997, 40, 260-273.

    2. Frazer, A.; Hensler, J. G. In Basic Neurochemistry; Siegel G. J.;

    Agranoff, B. W.; Alber, R. W.; et al, Eds.; Raven Press: New York,

    1993.

    3. Richelson, E. M. Clin. Proc. 1994, 69, 1069.

    4. a) Owens, M. J.; Nemeroff, C. B. Clin. Chem. 1994, 40, 288-295. b)

    Frazer, A. J. Clin. Psychiatry 1997, 6, 9-25. c) Hyttel, J. Int. Clin.

    Psychopharmacol. 1994, 1, 19-26.d) Staley, J. K.; Malison, R. T.;

    Innis, R. B. Biol. Psychiatry 1998, 44, 534-549. e) Oh, S. J.; Ha, H.-

    J.; Chi, D. Y.; Lee, H. K. Current Med. Chem. 2001, 8, 999-1034.

    5. Rothman, R. B.; Baumann, M. H. Pharmacol Ther. 2002, 95, 73-88.

    6. Szabo, Z.; Kao, P. F.; Scheffel, U.; Suehiro, M.; Mathews, W. B.;

    Ravert, H. T.; Musachio, J. L.; Marenco, S.; Kim, S. E.; Ricaurte, G.

    A.; Wong, D. F.; Wagner, H. N., Jr.; Dannals, R. F. Synapse 1995,

    20, 37-43.

    7. a) Suehiro, M.; Musachio, J. L.; Dannals, R. F.; Mathews, W. B.;

    Ravert, H. T.; Scheffel, U.; Wagner, H. N. Nucl. Med. Biol. 1995, 22,

    543-545. b) Suehiro, M.; Scheffel, U.; Dannals, R. F.; Ravert, H. T.;

    Ricaurte, G. A.; Wagner, H. N. J. Nucl. Med. 1993, 34, 120-127. c)

    Elfving, B.; Bjornholm, B.; Ebert, B.; Knudsen, G. M. Synapse 2001,

    41, 203-211.

    8. Maryanoff, E. M.; Vaught, J. L.; Shank, R. P.; McComsey, D. F.;

    Costanzo, M. J.; Nortey, S. O. J. Med. Chem. 1990, 33, 2793-2797.

  • 27

    9. a) Szabo, Z.; Scheffel, U.; Mathews, W. B.; Ravert, H. T.; Szabo, K.;

    Kraut, M.; Palmon, S.; Ricaurte, G. A.; Dannals, R. F. J. Cereb.

    Blood Flow Metab. 1999, 19, 967-981. b) Szabo, Z.; Mohamadiyeh,

    M.; Scheffel, U.; Matthews, W. B.; Ravert, H. T.; Szabo, K.; Palmon,

    S.; Dannals, R. F. J. Nucl. Med. 1996, 37, 125.

    10. Parsey, R. V.; Kegeles, L. S.; Hwang, D. R.; Simpson, N.; Abi-

    Dargham, A.; Mawlawi, O.; Slifstein, M.; Van Heertum, R. L.;

    Mann, J. J.; Laruelle, M. J. Nucl. Med. 2000, 41, 1465-1477.

    11. a) Ricaurte, G. A.; McCann, U. D.; Szabo, Z.; Scheffel, U. Toxicol

    Lett. 2000, 112-113, 143-146. b) McCann, U. D.; Szabo, Z.; Scheffel,

    U.; Dannals, R. F.; Ricaurte, G. A. Lancet 1998, 352, 1433-1437.

    12. a) Chumpradit, S.; Kung, M.-P.; Panyachotipun, C.; Prapansiri, V.;

    Foulon, C.; Brooks, B. P.; Szabo, S. A.; Tejani-Butt, S.; Frazer, A.;

    Kung, H. F. J. Med. Chem. 1992, 35, 4492-4497. b) Soudijn, W.; van

    Wijngaarden, I. Serotonin Receptors and Their Ligands; Elsevier

    Science: New York, 1997; pp 327-361. c) Suehiro, M.; Scheffel, U.

    A.; Ravert, H. T.; Ricaurte, G. A.; Hatzidimitriou, G.; Dannals, R.

    F.; Bogeso, K. P.; Wagner, H. N. Nucl. Med. Biol. 1994, 21, 1083-

    1091. d) Kung, M.-P.; Chumpradit, S.; Billings, J. J.; Kung, H. F.

    Life Sci. 1992, 51, 95-106.

    13. a) Biegon, A.; Mathis, C. A.; Hanrahan, S. M.; Jagust, W. J. Brain

    Res. 1993, 619, 236-246. b) Mathis, C. A.; Taylor, S. E.; Biegon, A.;

    Enas, J. D. Brain Res. 1993, 619, 229-235. c) Mathis, C. A.; Biegon,

    A.; Taylor, S. E.; Enas, J. D.; Hanrahan, S. M. Eur. J. Pharmacol.

    1992, 210, 103-104.

    14. Jagust, W. J.; Eberling, J. L.; Biegon, A.; Taylor, S. E.; Van-

    Brocklin, H. F.; Jordan, S.; Hanrahan, S. M.; Roberts, J. A.; Brennan,

    K. M.; Mathis, C. A. J. Nucl. Med. 1996, 37, 1207-1214.

  • 28

    15. Ferris, R. M.; Brieaddy, L.; Mehta, N.; Hollingsworth, E.; Rigdon,

    G.; Wang, C.; Soroko, F.; Wastila, W.; Cooper, B. J. Pharm.

    Pharmacol. 1995, 47, 775-781.

    16. Oya, S.; Kung, M. P.; Acton, P. D.; Mu, M.; Hou, C.; Kung, H. J.

    Med. Chem. 1999, 42, 333-335.

    17. Oya, S.; Choi, S. R.; Hou, C.; Mu, M.; Kung, M. P.; Acton, P. D.;

    Siciliano, M.; Kung, H. F. Nucl. Med. Biol. 2000, 27, 249-254.

    18. Zhuang, Z. P.; Choi, S. R.; Hou, C.; Mu, M.; Kung, M. P.; Acton, P.

    D.; Kung, H. F. Nucl. Med. Biol. 2000, 27, 169-175.

    19. a) Acton, P. D.; Kung, M. P.; Mu, M.; Plossl, K.; Hou, C.; Siciliano,

    M.; Oya, S.; Kung, H. F. Eur. J. Nucl. Med. 1999, 26, 854-861. b)

    Kung, M. P.; Hou, C.; Oya, S.; Mu, M.; Acton, P. D.; Kung, H. F.

    Eur. J. Nucl. Med. 1999, 26, 844-853.

    20. a) Choi, S. R.; Hou, C.; Oya, S.; Mu, M.; Kung, M. P.; Siciliano, M.;

    Acton, P. D.; Kung, H. F. Synapse 2000, 38, 403-412.b) Acton, P.

    D.; Choi, S. R.; Hou, C.; Plossl, K.; Kung, H. F. J. Nucl. Med. 2001,

    42, 1556-1562.

    21. a) Lee, B. S.; Chu, S.; Lee, K. C.; Lee, B.-S.; Chi, D. Y.; Kim, S. E.;

    Choe, Y. S.; Song, Y. S.; Jin, C. Bioorg. Med. Chem. 2003, 11,

    4949-4958. b) Lee, B. S.; Chu, S.; Lee, B.-S.; Chi, D. Y.; Song, Y.

    S.; Jin, C. Bioorg. Med. Chem. Lett. 2002, 12, 811-815. c) In, M. Y.;

    Chi, D. Y.; Choi, S. J.; Park, K. B.; Cho, C. G. Bull. Korean Chem.

    Soc. 2002, 23, 1439-1444. d) Lee, B. S.; Chu S.; Lee, I. Y.; Lee, B.-

    S.; Song, C. E.; Chi, D. Y. Bull. Korean Chem. Soc. 2000, 21, 860-

    866. e) Lee, B. S.; Chu, S.; Lee, B. C.; Chi, D. Y.; Choe, Y. S.;

    Jeong, K. J.; Jin, C. Bioorg. Med. Chem. Lett. 2000, 10, 1559-1562.

    22. a) Batcho, A. D.; Leimgruber, W. Org. Synth. 1985, 63, 214-220. b)

    Clark, R. D.; Repke, D. B. Heterocycles 1984, 22, 195-221.

  • 29

    23. Hama, Y.; Nobuhara, Y.; Aso, Y.; Otsubo, T.; Ogura, F. Bull. Chem.

    Soc. Jpn. 1988, 61, 1683-1686.

    24. Clayton, S. C.; Regan, A. C. Tetrahedron Lett. 1993, 34, 7493-7496.

    25. Vogel, A. I. .Vogel’s Text book of practical organic chemistry, 5th

    ed.; revised by Furiss, B. S.; Hannaford, A. J.; Smith, P. W. G.;

    Tatchell, A. R. Longman Group UK Limited: Harlow, England,

    1989.

    26. Newman, M. S.; Karnes, H. A. J. Org. Chem. 1966, 31, 3980-3984.

  • 30

    NMR spectra

    2-Methyl-3-nitrophenyl N,N-Dimethylthionocarbamate (13). 1H NMR (200 MHz, CDCl3)

    13C NMR (50 MHz, CDCl3)

    O

    NO2

    S

    Me2N

  • 31

    2-Methyl-3-nitrophenyl N,N-Dimethylthiocarbamate (14). 1H NMR (200 MHz, CDCl3)

    13C NMR (50 MHz, CDCl3)

    S

    NO2

    O

    Me2N

  • 32

    2-Methyl-3-nitrothiophenol (8). 1H NMR (200 MHz, CDCl3)

    13C NMR (50 MHz, CDCl3)

    SH

    NO2

  • 33

    1-(5-Iodopyridin-2-ylthio)-2-methyl-3-nitrobenzene (6). 1H NMR (200 MHz, CDCl3)

    13C NMR (50 MHz, CDCl3)

    S

    NO2

    N

    I

  • 34

    4-(5-Iodopyridin-2-ylthio)indole (16). 1H NMR (200 MHz, DMSO-d6)

    13C NMR (50 MHz, DMSO-d6)

    SN

    I

    NH

  • 35

    3-Acetyl-4-(5-iodopyridin-2-ylthio)indole (17).

    1H NMR (200 MHz, CDCl3)

    13C NMR (50 MHz, CDCl3)

    SN

    I

    NH

    O

  • 36

    3-(4-Iodopyridin-2-yl)-4-aminobenzothiophene (18). 1H NMR (400 MHz, CDCl3)

    13C NMR (100 MHz, CDCl3)

    NH2

    S N

    I

  • 37

    3-(4-Iodopyridin-2yl)-4-aminothiophene (18)

    2D-HETCOR. in CDCl3

    NH2

    S N

    I

  • 38

    3-Formyl-4-(5-iodopyridin-2-ylthio)indole (19). 1H NMR (400 MHz, DMSO-d6)

    13C NMR (100 MHz, DMSO-d6)

    SN

    I

    NH

    CHO

  • 39

    3-(Aminomethyl)-4-(5-iodopyridin-2-ylthio)indole (20) 1H NMR (400 MHz, DMSO-d6)

    13C NMR (100 MHz, DMSO-d6)

    SN

    I

    NH

    NH2

  • 40

    3-(Hydroxymethyl)-4-(5-iodopyridin-2-ylthio)indole (21) 1H NMR (400 MHz, DMSO-d6)

    13C NMR (100 MHz, DMSO-d6)

    SN

    I

    NH

    OH

  • 41

    PART 2

    Structural Modification of Nitric Oxide Inhibitors

  • 42

    Introduction

    Nitric oxide or nitrogen monoxide, a small free radical, is formed

    by nitric oxide synthases (NOS; EC 1.14.13.39). In the presence of

    oxygen and NADPH, NOS catalyses five-electron oxidation of the

    terminal guanidino nitrogen atoms of L-arginine to generate L-citrulline

    and NO (Scheme 1)1 using flavin adenine dinucleotide (FAD), flavin

    mononucleotide (FMN), heme, and tetrahydrobiopterin (BH4) as

    cofactors.2 Functional NOS has two bidomain structures. The reductase

    domain of NOS at a C-terminus contains the binding sites for FAD, FMN,

    NADPH and calmodulin, and a N-terminal oxygenase domain contains the

    binding sites for heme, BH4 and L-arginine. The N-terminal oxygenase

    domain is linked by a calmodulin recognition site to the C-terminal

    reductase domain. Native NOS is a homodimer, which requires heme for

    dimerization of monomers and full NOS activity.3

    NH

    COOH3N

    NH2H2NNADPH+H+ NADP+

    O2 H2O

    NH

    COOH3N

    HNH2N OH

    0.5 NADPH+H+

    O2BH4

    NADP +

    H2O

    NO

    NH

    COOH3N

    OH2N

    +

    L-Arginine NG-Hydroxy-L-arginine Nitric oxide L-C itrulline Scheme 1. The biosynthesis of nitric oxide.

    Three different NOS isoforms have been characterized in

    mammalian tissues.3 Two constitutively expressed NOS isoforms which

    are activated by stimulation dependent Ca2+ entry, are present mainly in

    brain (neuronal NOS; nNOS or Type I NOS) and endothelial cell

    (endothelial NOS; eNOS or Type III NOS). Nitric oxide produced by

  • 43

    nNOS functions mainly in neurotransmission while the NO produced by

    eNOS functions in the maintenance of normal vascular homeostasis such

    as regulation of blood pressure and prevention of platelet and leukocyte

    adhesion and activation. A third isoform, cytokine-inducible and Ca2+-

    independent NOS (inducible NOS; iNOS or Type II NOS) is expressed in

    macrophages, neutrophils, hepatocytes and other cells. The expression of

    iNOS is induced after stimulation with lipopolysaccharide (LPS) and pro-

    inflammatory cytokines e.g. interleukin (IL), tumor nrcrosis factor-α

    (TNF-α), and interferons (IFN).4 High amount of nitric oxide produced by

    iNOS functions mainly in pathogen killing processes, where its toxicity

    can be due to a combination of effects, including inhibition of target cell

    respiration and cell division, reaction with redox active catalytic iron

    centers, and indirect cytotoxicity through formation of toxic oxidants as

    well as nitrating/nitrosating species.5 Moreover, BH4, an essential cofactor

    for the activation of all isoforms of NOS, is synthesized from GTP via

    sequential enzyme reactions catalyzed by GTPcyclohydrolase I (GTPCH),

    6-pyruvoyl-tetrahydropterin synthase, and sepiapterin reductase. It was

    reported that cytokine-induced NO production requires GTPCH activation

    in cardiac myocytes.6 Also, increased BH4 levels in murine fibroblasts,7

    cardiac myocytes,8 and endothelial cells9 indicate that the availability of

    the cofactor regulates NOS activity.

    Nitric oxide (NO) and other free radicals have been implicated in

    the pathophysiology of ischemic neuronal death.10 NO is an important

    signaling molecule in normal synaptic transmission but can be a

    neurotoxin under pathological conditions. Increases in NO generation,

    NOS mRNA, and protein were reported in animal models of ischemia,11

    and NOS inhibitors protected neurons in these animal models.12 The

  • 44

    mechanism by which NO contributes to ischemic neuronal death, either

    through necrosis or apoptosis, is not known. However, NO-mediated

    hydrolytic cleavage of poly(ADP-ribose)-polymerase, one of the key

    substrates for activated cysteine protease,13 suggests that NO plays a role

    in apoptotic cell death.

    Figure 1. Current major approaches A-D for NO inhibition.

    There are many approaches to inhibit NO production, not only at

    transcription level but also at postranscription level, as shown in Figure 1.

    NO production is blocked at DNA or mRNA level (approach A) related to

  • 45

    the production of cofactors for iNOS activity.14 In approach B, GTP

    derivatives derived from GTP or related compounds were tested for the

    inhibition of GTP cyclohydrolase (GTPCH),15 a rate-limiting enzyme for

    the conversion of GTP to BH4. BH4 is an essential cofactor for the

    production of NO16 and it also acts as glue to maintain NOS in dimeric

    active form.16,17 BH4 derivatives were also synthesized to inhibit

    dimerization of NOS (approach C).18 Recently, arginine mimetic

    compounds19 and other small molecules20 were reported to inhibit NO

    production by competitively blocking at substrate L-arginine binding site

    (approach D).

    The regulation of NO overproduction to prevent cell damage was

    interested widely. Although there are many ways to inhibit NO production,

    most of them also showed some side effect and toxicity.21 Recently, Cho

    and coworkers reported that treatment with N-acetyl-O-methyldopamine

    (NAMDA) (Chart 1), a metabolite of dopamine in CNS, significantly

    protected CA1 neurons in rat ischemic hippocampus and inhibited LPS-

    induced NO production in BV-2 microglia cells without direct inhibition

    of iNOS activity.22 More recently, it was demonstrated that NAMDA act

    as a neuroprotectant by repressing LPS-induced proinflammatory

    cytokines gene expression via a cAMP-dependent protein kinase pathway

    without preventing NF-κB nuclear translocation or its DNA binding

    activity in microglia cells.23 They suggested that NAMDA as a potent

    agent, which attenuated neuronal injury. Although, NAMDA is not toxic,

    it showed good protective activity only at high concentration (5 mM).

    Therefore, it was clear that structure modification was needed to improve

    its activity and to develop as a potent therapeutic agent.

  • 46

    H3CO

    HOHN

    O1 Chart 1 The structure of NAMDA 1.

    Figure 3 Strategies of structure modification

    The structure modification was made by homologation,

    cyclization and replacing of the acetamide group with other function

    groups as shown in Figure 3. It was considered that the restrict

    rotation of flexible carbon and changing of acetamide group would

    provide some improvement on its inhibitory activity. Herein, several

    compounds were synthesized and their biological effects were

    evaluated on NO production and cytotoxicity.

    H3CO

    HOHN

    OChanging of acetamide group

    Homologation

    Cyclization

  • 47

    Chemistry

    3-O-methyldopamine hydrochloride (4) was firstly synthesized by

    previously reported method.24 4-Hydroxy-3-methoxybenzyl alcohol (2)

    was converted to (4-hydroxy-3-methoxyphenyl)acetonitrile (3) with

    sodium cyanide, and then reduced to amine by hydrogenation using

    palladium on charcoal as a catalyst. The treatment of 4 with S-2-

    naphthylmethyl thioacetimidate hydrobromide25 in ethanol at room

    temperature gave acetamidine hydrobromide 5 in a high yield (Scheme 2).

    OH

    HO

    MeOCN

    HO

    MeO

    HO

    MeOHN

    NHHO

    MeO NH2.HCl

    2 3

    4

    a

    5

    b

    c

    Scheme 2. (a) NaCN, DMF, 130°C, 20 h, 60%; (b) H2, Pd/C, HCl, EtOH, rt, 12h,

    82%; (c) S-2-naphthylmethyl thioacetimidate hydrobromide, Et3N, EtOH, rt, 3 h,

    70%.

    In many attempt to synthesis of cyclic amide, the reactions were

    failed due to double bond formation by fast elimination at α,β position.

    The cyclic amine derivatives were, however, designed and synthesized as

    shown in Scheme 3. The nitrile 3 was hydrolyzed under basic condition to

    a carboxylic derivative 6. The coupling reaction of 6 with piperidine and

    pyrrolidine by 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide

  • 48

    hydrochloride (EDCI) yielded amide 7 and 8, respectively. Subsequently,

    the amide 7 and 8 were reduced using lithium aluminium hydride to

    corresponding amine 9 and 10.

    HO

    COOHMeO MeO

    HO

    R

    O

    N

    NMeO

    HO

    R

    N

    N

    3

    67, R =

    8, R =

    a b

    c

    9, R =

    10, R =

    Scheme 3. (a) KOH, EtOH, reflux, overnight, 74%; (b) piperidine or pyrrolidine,

    EDCI, DMAP, DMF, rt, 3 h, 75% for 7, 51% for 8; (c) LAH, THF, reflux, 2 h,

    69% for 9, 80% for 10.

    MeO

    HO

    O

    HMeO

    HO

    CN

    MeO

    HO

    NH

    OMeO

    HO

    NH2.HC l

    a b

    c

    11

    13 14

    12

    Scheme 4. (a) KOH, CH3CN, reflux, overnight, 83%; (b) H2, Pd/C, HCl, EtOH,

    rt, 12 h, %; (c) Ac2O, Et3N, CH2Cl2, rt, 30 min, 76%.

  • 49

    To obtain a homologous chain derivative (Scheme 4), vanillin (11)

    was refluxed with potassium hydroxide in acetonitrile and leaded to trans-

    cinamonitrile 12 as a major product. The catalytic hydrogenation of 12

    under acidic condition completely reduced both double bond and nitrile

    group to give amine 13 as an ammonium hydrochloride salt. The amide 14

    was consequently obtained by acetylation of 13 with acetic anhydride.

    Cyclized derivatives, tetrahydroisoquinoline, were next

    synthesized via the Pictet-Spengler synthesis26 as shown in Scheme 5. The

    treatment of 4 with formaldehyde gave 15 by one-pot synthesis. N-

    formylation of 15 with trimethyl orthoformate was performed in a sealed

    tube under N2 atmosphere and need to add concentrated hydrochloride in a

    catalytic amount to yield formamide 16. Lastly, tetrahydroisoquinoline 15

    was simply treated with benzoic anhydride and lead to benzamide 17.

    MeO

    HONH

    MeO

    HON H

    O

    MeO

    HON

    O

    .HCl

    4a

    b c

    15

    16 17

    Scheme 5. (a) HCHO, H2O, HCl, 70°C, 1 h, 70%; (b) trimethylorthoformate,

    conc.HCl (cat.), reflux, overnight, 90%; (c) Bz2O, Et3N, CH2Cl2, rt, 30 min, 65%

  • 50

    Biological Results and Discussion.

    Table 1 Nitrite and LDH production assay: BV2 Cell (2.5x104 cells/well)a

    % of LPS alone Compound

    Nitrite a LDH cytotoxicity assay a

    5 96.9 96.4

    7 90.3 81.1

    8 56.1 68.8

    9 85.9 78.2

    10 84.4 83.7

    12 54.0 100.4

    14 77.5 98.6

    15 16.5 75.4

    16 128.8 108.1

    17 84.9 104.7

    NAMDA (1) 84.0 73.4

    LPS 100.0 100.0

    control 37.8 77.2 aEach assay was treated at 100 µM concentration of tested compound.

  • 51

    HO

    MeOHN

    NHHO

    MeO N

    OHO

    MeO N

    O

    HO

    MeO N

    HO

    MeO N MeO

    HO

    CN

    MeO

    HO

    NH

    OMeO

    HONH.HC l

    MeO

    HON H

    O

    MeO

    HON

    O

    5 87

    9 10 12

    14 15 16

    17 Chart 2 The structure of tested compounds.

    The inhibitory activity and cytotoxicity of 11 compounds including NAMDA on NO production induced by lipopolysaccharide (LPS) in microgial (BV2) were shown in Table 1. Those compounds (Chart 2) were modified structurally at amide group or chain but remained catechol moiety untouched. Among open-chain compounds, three compounds 8, 12, 14 showed significantly more potent activity than that of NAMDA and all did not show cytotoxicity in LDH assay. Both amine 9, 10 and acetamidine 5 showed similar potency to NAMDA maybe because of their polarity, whereas less polar compound such as amide 8 and cinamonitrile 12 are more potent. Interestingly, the homologous compound 14 showed some improvement of inhibitory activity. This result indicated that less polar function group trended to show better potency. Next, three tetrahydroisoquinolines were tested. Compound 15 showed excellent activity, which could reduce NO production lower than that of control sample, whereas amide 16 increased NO production. Among all tested compounds, tetrahydroisoquinoline 15 showed the best result and could successfully inhibit NO

  • 52

    production induced by LPS in microgial cell at 100 µM concentration. This compound can be a new lead compound for development of NO inhibitor due to its much higher activity and non-cytotoxicity. The extensive study including mechanism and target enzyme would, however, be investigated and confirmed further.

  • 53

    Conclusions

    Ten compounds were synthesized and evaluated its biological

    activity for NO production. The structural modification was made by

    homologation, cyclization and changing acetamide group to other

    functional groups, acetamidine, amides, cyclic amines and cinamonitrile.

    Among all compounds, four compounds showed some improvement of

    inhibitory activity without increasing cytotoxicity. Compound 15

    exhibited strikingly as the most potent NO reducing agent and more

    significantly potent than the lead compound, NAMDA.

  • 54

    Experimental Section

    4-Hydroxy-3-methoxyphenylacetonitrile (3). To a solution of 4-

    hydroxy-3-methoxybenzly alcohol (2) (20 g, 0.13 mol) in DMF (300 mL)

    was added potassium cyanide (0.16 mol, 10 g) and then set the

    temperature at 130 °C under N2. After 20 h, reaction mixture was cooled

    to room temperature and quenched by water. Brine was added and

    extracted with chloroform. Organic layer was washed two times more with

    Brine and water. Chloroform was evaporated, and dark brown mixture was

    placed to vacuum distillation apparatus. Vacuum distillation was proceed

    at 0.6 torr and 120 °C. 3 (12.7 g, 60%) was obtained as a white solid: 1H

    NMR (CDCl3, 200 MHz) δ 6.88-6.73 (m, 3H), 5.89 (s, 1H), 3.84 (s, 3H),

    3.64 (s, 3H); 13C NMR (CDCl3, 50 MHz) 146.9, 145.3, 121.4, 120.7,

    144.7, 110.4, 55.8, 22.9. CAS No. 4468-59-1

    3-O-Methyldopamine hydrochloride (4). To a solution of 3 (6.0 g, 37

    mmol) in ethanol (60 mL) was added 10% wet palladium charcoal (1.0 g).

    Round-bottom flask was sealed with septum and purged with H2 for 2 min,

    then conc. HCl (6 mL) was added. Mixture was stirred for 12 h at room

    temperature. Reaction mixture was filtered through celite and then solvent

    was evaporated. Crystallization in methanol and ethyl acetate gave 4 (6.2

    g, 82%) as a white solid: 1H NMR (DMSO-d6, 200 MHz)

    δ 8.92 (s, 1Η), 8.19 (br s, 2H), 6.81 (d, J = 1.6 Hz, 1H), 6.74 (d, J = 8.0 Hz,

    1H), 6.61 (dd, J = 8.0, 1.4 Hz, 1H), 3.75 (s, 3H), 3.05-2.83 (brm, 2H),

    2.81-2.66 (br m, 2H); 13C NMR (DMSO-d6, 50 MHz)

    δ 151.6, 149.3, 132.0, 124.8, 119.5, 116.9, 59.6, 44.1, 36.5. CAS No.

    1477-68-5.

  • 55

    N-[2-(4-Hydroxy-3-methoxyphenyl)ethyl]acetamidine hydrobromide

    (5) The suspension of 4 (0.50 g, 2.5 mmol) and triethylamine (0.7 mL) in

    EtOH (7.5 mL) was stirred at 0°C in an ice-salt bath. S-2-naphthylmethyl

    thioacetimidate hydrobromide25 was added and the reaction was allowed

    to stir at rt for 3 hr. The reaction was reduced in vacuo and residue was

    dissolved in water (20 mL). The aqueous solution was washed by ether

    (2x20 mL) and evaporated. The residue was crystallized in MeOH-ethyl

    acetate, giving 5 (0.50 g, 70%) as off-white solid. 1H NMR (DMSO-d6,

    200 MHz) δ 9.78 (brs, 1H), 9.24 (brs, 1H), 8.86 (brs, 1H), 8.75 (brs, 1H),

    6.92 (d, J = 1.4 Hz, 1H), 6.73 (d, J = 8.0 Hz, , 1H), 6.65 (dd, J =1.4 and

    8.0 Hz, 1H), 3.76 (s, 3H), 3.44 (t, J = 7.4 Hz, 2H), 2.74 (t, J = 7.4 Hz, 2H),

    2.16 (s, 3H); 13C NMR (DMSO-d6, 50 MHz) δ 164.5, 148.2, 145.9, 129.5,

    121.7, 116.1, 113.9, 56.4, 44.2, 33.7, 19.2; MS (CI) : 210, 208 (M+), 191,

    168, 151 (100), 138, 121.

    (4-Hydroxy-3-methoxyphenyl)acetic acid (6). The mixture of nitrile 3

    (4.00 g, 24.6 mmol) and KOH (10g, 178 mmol) in EtOH 200 mL was

    reflux for overnight. After cooling, the reaction was acidified by 5% HCl

    until pH

  • 56

    mixture was extracted with ethyl acetate (2x20 mL). The combined

    organic layers were washed by sat. NH4Cl, then dried over and evaporated.

    The residue was purified by column chromatography (EtOAc), yielding 7

    as solid (0.37 g, 75%). 1H NMR (CDCl3, 200 MHz) δ 6.84 (d, J = 7.6 Hz,

    1H), 6.80 (d, J = 1.8 Hz, 1H), 6.68 (dd, J = 1.6 and 8.0 Hz, 1H), 5.73 (s,

    1H), 3.86 (s, 3H), 3.64 (s, 2H), 3.55 (dd, J = 4.4 and 5.8 Hz, 2H), 3.38 (dd,

    J =5.4, 5.6Hz, 2H), 1.49-1.61 (m, 4H), 1.29 -1.40 (m, 2H); 13C NMR

    (CDCl3, 50 MHz) δ 169.7, 146.8, 144.5, 127.3, 121.5, 114.45, 111.1, 56.0,

    47.3, 43.0, 40.9, 26.3, 25.6, 24.5. CAS No. 53283-49-1.

    2-(4-Hydroxy-3-methoxy-phenyl)-1-pyrrolidin-1-yl-ethanone (8) The

    same procedure used as 7, 6 (0.85 g, 4.6 mmol), pyrrolidine (0.50 mL) in

    DMF (10 mL), EDCI (1.10 g, mmol) and DMAP (0.12 g) were used. The

    reaction gave 8 as off-white solid (0.55 g, 51%) 1H NMR (CDCl3, 200

    MHz) δ 6.85 (s, 1H), 6.82 (d, J = 7.6 Hz, 1H), 5.96 (s, 1H), 3.84 (s, 3H),

    3.56 (s, 2H), 3.39-3.50 (m, 4H), 1.78-1.93 (m, 4H); 13C NMR (CDCl3, 50

    MHz) δ 170.0, 146.9, 144.7, 126.6, 121.8, 114.4, 11.6, 56.0, 47.0, 46.1,

    42.0, 26.2, 24.4. CAS No. 131656-82-1.

    2-Methoxy-4-(2-piperidin-1-yl ethyl)phenol (9) To solution of 7 (0.37 g,

    1.49 mmol) in dry THF (7 mL), 1.0M LiAlH4 in THF (4 mL) was added.

    The reaction was refluxed for 2 hr. After cooling, ethyl acetate (1 mL),

    water (1 mL), and 2 M NaOH (2mL) were added respectively and stirred

    at rt for 30 min. The suspension was filtered through celite and washed by

    water and ethyl acetate. The filtrate extracted with ethyl acetate (2x10 mL).

    The organic layers were combined, dried with anhydrous NaSO4 and

    evaporated in vacuo, giving 9 (0.24 g, 69%) as off-white solid. 1H NMR

    (CDCl3, 200 MHz) δ 6.79 (d, J = 8.0 Hz, 1H), 6.68 (s, 1H), 6.66 (dd, J =

  • 57

    1.4 and 8.0 Hz, 1H), 3.84 (s, 3H), 2.78-2.71 (m, 2H), 2.57-2.49 (m, 6H),

    1.69-1.59 (m, 4H), 1.47-1.45 (m,2H); 13C NMR (CDCl3, 50 MHz) δ 146.9,

    144.2, 132.3, 121.2, 114.7, 111.5, 61.8, 55.9, 54.6, 33.2, 25.9, 24.5 –MS

    (FAB) 236 (M+H+), 234, 151, 98. HRMS (FAB) Calc. for C14H21NO2

    (M+H+) 236.1651. Found 236.1649.

    2-Methoxy-4-(2-pyrrolidin-1-yl ethyl)phenol (10) The same procedure

    used as 9, 8 (0.40 g, 1.70 mmol) in dry THF (8 mL), 1.0M LiAlH4 in THF

    (4 mL) were used. The reaction gave 8 as off-white solid (0.30 g, 80%). 1H NMR (CDCl3, 200 MHz) δ 7.4 (brs, 1H), 6.75 (d, J = 7.8 Hz, 1H), 6.67

    (s, 1H), 6.65 (dd, J = 2.0 and 7.6 Hz, 1H), 3.84, s, 3H), 2.60-2.83 (m, 8H),

    1.78-1.83 (m, 4H); 13C NMR (CDCl3, 50 MHz) δ 147.1, 144.4, 131.9,

    121.1, 114.9, 111.5, 58.8, 55.8, 54.3 (2C), 35.3, 23.47 (2C); MS (FAB)

    222 (M+H+, 100), 151, 84. HRMS (FAB) Calc. for C13H19NO2 (M+H+)

    222.1494. Found 222.1495.

    3-(4-Hydroxy-3-methoxyphenyl)acrylonitrile (12) The suspension of

    KOH (1.12 g, 20 mmol) in acetonitrile 20 mL was stirred at rt for 30 min.

    The solution of vanillin (1.52 g, 10 mmol) in acetonitrile 10 mL was

    added and the reaction was refluxed for overnight. After cooling, the

    reaction was added by 50 mL of water and then extracted by

    dichloromethane (2x40 mL). The combined organic layer was washed by

    5% HCl (40 mL) and brine. The residue was purified by column

    chromatography (50% ethyl acetate-hexane), giving 12 as off-white solid

    (1.12 g, 83%). 1H NMR (CDCl3, 200 MHz) δ 7.29 (d, J = 16.6 Hz, 1H),

    6.90-7.01 (m, 2H), 6.15 (s, 1H), 5.71 (d, J = 16.8 Hz, 1H), 3.92 (s, 3H); 13C NMR (CDCl3, 50 MHz) δ 150.6, 148.9, 147.1, 126.3, 122.6, 118.9,

    115.0, 108.9, 93.2, 56.2. CAS No. 71750-09-9.

  • 58

    4-(3-Amino-propyl)-2-methoxyphenol hydrochloride (13) The same

    procedure used as 4, 12 (0.42 g, 3.11 mmol), 10% palladium on charcoal

    (0.18 g), 95% EtOH (10 mL), conc.HCl 0.5 mL) were used. The reaction

    gave 13 as off-white solid (0.32 g, 58%). 1H NMR (DMSO-d6, 200 MHz)

    δ 8.0 (brs, 2H), 6.74 (d, J = 1.4 Hz, 1H), 6.68 (d, J = 8.0 Hz, 1H), 6.54 (dd,

    J = 1.8 and 8.0 Hz, 1H), 3.70 (s, 3H), 2.68 (t, J = 7.6 Hz, 2H), 2.50 (t, J =

    7.6 Hz, 1H), 1.75-1.83 (m, 2H); 13C NMR (DMSO-d6, 100 MHz) δ 148.1,

    145.3, 132.2, 121.0, 116.0, 113.1, 56.2, 38.8, 32.0, 29.5. CAS No.

    112798-57-9.

    N-[3-(4-Hydroxy-3-methoxyphenyl)propyl]acetamide (14) The same

    procedure used as 1, 13 (0.20 g,), acetic anhydride (92 mg, 0.92 mmol),

    dichloromethane (2 mL) and triethylamine (0.38 mL) were used. The

    reaction gave 14 as colorless liquid (0.16 g, 76%). 1H NMR (CDCl3, 200

    MHz) δ 6.81 (d, J = 7.6 Hz, 1H), 6.65 (d, J = 1.8 Hz, 1H), 6.62 (dd, J =

    1.8 and 8.0 Hz, 1H), 6.41 (brs, 1H), 6.19 (brs, 1H), 3.83 (s, 3H), 3.24 (dt, J

    = 6.2 and 6.8 Hz, 2H), 2.55 (t, J = 7.6 Hz, 2H), 1.94 (s, 3H), 1.70-1.85 (m,

    2H); 13C NMR (CDCl3, 50 MHz) δ 170.7, 146.8, 144.0, 133.4, 120.8,

    114.6, 111.3, 56.0, 39.4, 33.0, 31.4, 23.3; MS (FAB) 224 (M+H+, 100),

    182, 164, 136. HRMS (FAB) Calc. for C12H17NO3 (M+H+) 224.1287.

    Found 224.1283.

    6-Methoxy-7-hydroxy-1,2,3,4-tetrahydroisoquinoline hydrochloride

    (15) To solution of 4 (1.0 g, 4.91 mmol) in 1N HCl was added 36%

    formaldehyde (2.5 mL) under N2 atmosphere and heated at 70 °C for 1 h.

    The solvent was removed by evaporator. The residue was crystallized in

    EtOH-ether to yield 15 as pale yellow solid (0.74 g, 70%). 1H NMR

  • 59

    (DMSO-d6, 200 MHz) δ 9.39 (br, 2H), 9.08 (br, 1H), 6.75 (s, 1H), 6.60 (s,

    1H), 4.07 (brs, 2H), 3.74 (s, 3H), 3.27-3.30 (brm, 2H), 2.88 (t, J= 9.8 Hz,

    2H); 13C NMR (DMSO-d6, 50 MHz) δ 146.7, 145.2, 128.6, 125.6, 113.7,

    113.3, 56.4, 47.7, 44.1, 28.7. CAS No. 1078-26-8.

    7-Hydroxy-6-methoxy-3,4-dihydro-1H-isoquinoline-2-carbaldehyde

    (16) Trimethyl orthoformate (2 mL) and conc.HCl (2 drops) were added to

    pressure tube which contains 15 (260 mg, 1.2 mmol) under N2. After tube

    was tightly capped, mixture had been refluxed for overnight. Mixture was

    cooled to room temperature and remaining trimethyl orthoformate was

    evaporated. Crude organic was separated by flash column chromatography

    (2% methanol-dichloromethane). 16 (224 mg, 90%) was obtained as a off-

    white solid: 1H NMR (CDCl3, 200 MHz, a mixture of two isomers) δ 8.23

    (s, 1H), 8.17 (s, 1H), 6.67 (s, 1H), 6.64 (s, 1H), 6.60 (s, 1H), 6.58 (s, 1H),

    6.11 (s, 1H), 4.57 (s, 1H), 4.42 (s, 1H), 3.86 (s, 3H), 3.85 (s, 3H), 3.76 (t, J

    = 6.4 Hz, 2H), 3.61 (t, J = 5.8 Hz, 2H), 2.74-2.84 (m, 6H); 13C NMR

    (CDCl3, 50 MHz, a mixture of two isomers) δ 161.6, 161.2, 145.6, 144.7,

    144.4, 125.4, 124.6, 124.1, 112.2, 111.6, 111.1, 110.9, 55.9, 46.9, 43.4,

    41.8, 38.1, 29.3, 27.5; MS (EI) 207 (M+, 100), 192, 178, 163, 150, 135,

    107, 91, 77, 67, 51. CAS No. 36646-95-4.

    (7-Hydroxy-6-methoxy-3,4-dihydro-1H-isoquinolin-2-yl)-phenyl-

    methanone (17) To a solution of 15 (215 mg, 1.0 mmol) in

    dichloromethane (8 mL) was added benzoic anhydride (0.9 mmol, 205

    mg) and triethylamine (0.45 mL) slowly at room temperature. Reaction

    mixture was stirred for 30 min. Reaction was diluted by dichloromethane

    and then washed with saturated ammonium chloride solution and water.

    Organic layer was dried over sodium sulfate. After then, solvent was

  • 60

    evaporated and flash column chromatography gave 17 (184 mg, 65%) as a

    yellow solid: 1H NMR (CDCl3, 200 MHz, a mixture of two isomers) δ

    7.43 (s, 5H), 6.45-6.74 (br(m), 2H), 5.72 (brs, 1H), 4.77 (brs, 1H), 4.46

    (brs, 1H), 3.94 (brs, 1H), 3.86 (s, 3H), 3.60 (brs, 1H), 2.79 (brs, 2H); 13C

    NMR (CDCl3, 100 MHz, a mixture of two isomers) δ 170.9, 170.4, 145.5,

    144.5, 144.2, 136.1, 129.7, 128.5, 127.1, 126.8, 125.8, 125.3, 124.9, 112.4,

    111.5, 110.9, 110.7, 55.9, 49.3, 45.4, 44.3, 40.6, 29.2, 27.8; MS (EI) 283

    (M+), 268, 178, 163, 150, 131, 105 (100), 77, 51; HRMS (FAB) Calc. for

    C17H17NO3 (M+H+) 284.1287. Found 284.1288.

  • 61

    References

    1. Marletta, M. A. J. Biol. Chem. 1993, 268, 12231-12234.

    2. MacMicking, J.; Xie, Q. W.; Nathan, C. Annu. Rev. Immunol. 1997,

    15, 323-350.

    3. Alderton, W. K.; Cooper, C. E.; Knowles, R. G. Biochem. J. 2001,

    357, 593-615.

    4. a) Jacobs, A. T.; Ignarro, L. J. J. Biol. Chem. 2001, 276, 47950-

    47957. b) Nathan, C. FASEB J. 1992, 6, 3051-3064.

    5. a) Coleman, J. W. Int. Immunopharmacol. 2001, 1, 1397-1406. b)

    Eiserich, J. P.; Patel, R. P.; O’Donnell V. B. Molec. Aspects Med.

    1998, 19, 221-357.

    6. Oddis, C. V.; Finkel, M. S. Am. J. Physiol. 1996, 270, H1864–

    H1868.

    7. Werner-Felmayer, G.; Werner, E. R.; Fuchs, D.; Hausen, A.;

    Reibnegger, G.; Wachter, H. J. Exp. Med. 1990, 172, 1599 –1607.

    8. Kasai, K.; Hattori, Y.; Banba, N.; Hattori, S.; Motohashi, S.;

    Shimoda, S.; Nakanishi, N.; Gross, S. S. Am. J. Physiol. 1997, 273,

    H665–H672.

    9. a) Werner-Felmayer, G.; Werne,r E. R.; Fuchs, D.; Hausen, A.;

    Reibnegger, G.; Schmidt, K.; Weiss, G.; Wachter, H. J. Biol. Chem.

    1993, 268, 1842–1846. b) Rosenkranz-Weiss, P.; Sessa, W. C.;

    Milstien, S.; Kaufman, S.; Watson, C. A.; Pober, J. S. J. Clin. Invest.

    1994, 93, 2236 –2243.

    10. a) Patt, A.; Harken, A. H.; Burton, L. K.; Rodell T. C.; Piermattei D.;

    Schorr, J.; Parker, N. B.; Berger, E. M.; Horesh, I. R.; Terada, L. S.;

    Linas, S. L.; Cheronis, J. C.; Repine, J.E. J. Clin. Invest. 1988, 81,

  • 62

    1556 –1562. b) Kader, A.; Frazzini, V. I.; Solomon, R. A.; Trifiletti,

    R. R. Stroke 1993, 24, 1709 –1716.

    11. a) Zhang, Z. G.; Chopp, M.; Gautam, S.; Zaloga, C.; Zhang, L.;

    Schmidt, H. H.; Pollock, J. S.; Forstermann, U. Brain Res. 1994,

    654, 85–95. b) Iadecola, C. Trends Neurosci. 1997, 20, 132–139.

    12. a) Buisson, A.; Plotkine, M.; Boulu, R. G. Br. J. Pharmacol. 1992,

    106, 766 –767. b) Hamada, Y.; Hayakawa, T.; Hattori, H.; Mikawa,

    H. Pediatr. Res. 1994, 35, 10 –14. c) Huang, Z.; Huang, P. L.;

    Panahian, N.; Dalkara, T.; Fishman, M. C.; Moskowitz, M. A.

    Science 1994, 265, 1883–1885.

    13. a) Bonfoco, E.; Zhivotovsky, B.; Rossi, A. D.; Aguilar-Santelises,

    M.; Orrenius, S.; Lipton, S. A.; Nicotera, P. Neuro. Report 1996, 8,

    273–276. b) Messmer, U. K.; Reimer, D. M.; Reed, J. C.; Brune, B.

    FEBS Lett. 1996, 384, 162–166.

    14. Fujiwara, N.; Okado, A; Seo, H. G.; Fujii, J.; Kondo, K; Taniguchi,

    N. FEBS Letter. 1996, 395, 299-303.

    15. Xie, L.; Smith, J. A.; Gross, S. S. J. Biol. Chem. 1998, 273, 21091-

    21098.

    16. Kinoshita, H.; Tsutsui, M.; Milstien, S.; Katusic, Z. S. Prog. Neuro.

    Biol. 1997, 52, 295-302. 17. Frohlich, L. G.; Kotsonis, P.; Traub, H.; Taghavi-Moghadam, S.; Al-Masoudi,

    N.; Hofmann, H.; Strobel, H.; Matter, H.; Pfleiderer, W.; Schmidt, H. H. H.

    W.; J. Med. Chem. 1999, 42, 4108-4121

    18. a) Werner, E.R.; Pitters, E.; Schmidt, K.; Wachter, H.; Werner-

    Felmayer, G.; Maye,r B. Biochem. J. 1996, 320, 193-6. b) Bommel,

    H. M.; Reif, A.; Frohlich, L. G.; Frey, A.; Hofmann, H.; Marecak, D.

    M.; Groehn, V.; Kotsonis, P.; La, M.; Koster, S.; Meinecke, M.;

    Bernhardt, M.; Weeger, M.; Ghisla, S.; Prestwich, G. D.; Pfleiderer,

    W.; Schmidt, H. H. J. Biol. Chem. 1998, 273, 33142-33149.

  • 63

    19. a) Huang, H.; Martasek, P.; Roman, L. J.; Silverman, R. B.; J. Med.

    Chem. 2000, 43, 2938-2945. b) Huang, H.; Martasek, P.; Roman, L.

    J.; Masters, B. S. S.; Silverman, R. B.; J. Med. Chem. 1999, 42,

    3147-3153. c) Collins, J. L.; Shearer, B. G.; Oplinger, J. A.; Lee, S.;

    Garvey, E. P.; Salter, M.; Duffy, C.; Burnette, T. C.; Furfine, E. S.; J.

    Med. Chem. 1998, 41, 2858-2871.

    20. a) Schumann, P.; Collot, V.; Hommet, Y.; Gsell, W.; Dauphin, F.;

    Sopkova, J.; MacKenzie, E. T.; Duval, D.; Boulouard, M.; Rault, S.

    Bioorg. Med. Chem. Lett. 2001, 11, 1153-6. b) Moormann, A. E.;

    Metz, S.; Toth, M. V.; Moore, W. M.; Jerome, G.; Kornmeier, C.;

    Manning, P.; Hansen, D. W. Jr.; Pitzele, B. S.; Webber, R. K. Bioorg.

    Med. Chem. Lett. 2001, 11, 2651-3. c) Hagmann, W. K.; Caldwell, C.

    G.; Chen, P.; Durette, P. L.; Esser, C. K.; Lanza, T. J.; Kopka, I. E.;

    Guthikonda, R.; Shah, S. K.; MacCoss, M.; Chabin, R. M.; Fletcher,

    D.; Grant, S. K.; Green, B. G.; Humes, J. L.; Kelly, T. M.; Luell, S.;

    Meurer, R.; Moore, V.; Pacholok, S. G.; Pavia, T.; Williams, H. R.;

    Wong, K. K. Bioorg. Med. Chem. Lett. 2000, 10, 1975-8.

    21. Southan, G. J.; Szabo, C. Biochem. Pharmacol. 1996, 51, 383-94.

    22. Cho, S.; Volpe, B. T.; Bae, Y.; Hwang, O.; Choi, H. J.; Gal, J.; Park,

    L. C. H.; Chu, C. K.; Du, J.; Joh, T. H. J. Neurosci. 1999, 19, 878–

    889.

    23. Cho, S.; Kim, Y.; Cruz, M. O.; Park, E. M.; Chu, C. K.; Song, G. Y.;

    Joh, T.H. GLIA 2001, 33, 324–333.

    24. Schwartz, M. A; Zoda, M.; Vishnuvajjala, B.; Mami, I. J. Org. Chem.

    1976; 41, 2502-2503.

    25. Shearer, B.G.; Oplinger, J. A.; Lee, S. Tetrahedron Lett. 1997, 38,

    179-182.

    26. Whaley, W. M.; Govindachari, T. R. Org. React. 1951, 6, 151.

  • 64

  • 65

    NMR Spectra

    N-[2-(4-Hydroxy-3-methoxyphenyl)ethyl]acetamidine hydrobromide

    (5) 1H NMR (DMSO-d6, 200 MHz)

    13C NMR (DMSO-d6, 50 MHz)

    HO

    MeOHN

    NH

  • 66

    2-(4-Hydroxy-3-methoxy-phenyl)-1-piperidin-1-yl-ethanone (7) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    HO

    MeO N

    O

  • 67

    2-(4-Hydroxy-3-methoxy-phenyl)-1-pyrrolidin-1-yl-ethanone (8) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    HO

    MeO N

    O

  • 68

    2-Methoxy-4-(2-piperidin-1-yl ethyl)phenol (9) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    HO

    MeO N

  • 69

    2-Methoxy-4-(2-pyrrolidin-1-yl ethyl)phenol (10) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    HO

    MeO N

  • 70

    3-(4-Hydroxy-3-methoxyphenyl)acrylonitrile (12) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    MeO

    HO

    CN

  • 71

    N-[3-(4-Hydroxy-3-methoxyphenyl)propyl]acetamide (14) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    MeO

    HO

    NH

    O

  • 72

    6-Methoxy-7-hydroxy-1,2,3,4-tetrahydroisoquinoline hydrochloride

    (15) 1H NMR (DMSO-d6, 200 MHz)

    13C NMR (DMSO-d6, 50 MHz)

    MeO

    HONH.HCl

  • 73

    7-Hydroxy-6-methoxy-3,4-dihydro-1H-isoquinoline-2-carbaldehyde

    (16) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    MeO

    HON H

    O

  • 74

    (7-Hydroxy-6-methoxy-3,4-dihydro-1H-isoquinolin-2-yl)-phenyl-

    methanone (17) 1H NMR (CDCl3, 200 MHz)

    13C NMR (CDCl3, 50 MHz)

    MeO

    HON

    O

  • 75

    PART 3

    Electrophilic Aromatic Addition Reaction: AdEAr

  • 76

    Introduction

    Although a wide variety of electrophilic species can attack

    aromatic rings and effect substitution, a single broad mechanism (Figure

    1) encompasses the large majority of electrophilic aromatic substitution

    reactions through the reversible formation of π and σ complexes.1 In this

    mechanism, a complexation of the electrophile with the π-electron system

    of the aromatic ring is the first step. This species, called the π-complex,

    may or may not be involved directly in the substitution mechanism. π-

    Complex formation is, in general, rapidly reversible, and in many cases

    the equilibrium constant is small. The π-complex is a donor-acceptor type

    complex, with the π electrons of the aromatic ring donating electron

    density to the electrophile. No position selectivity is associated with the π-

    complex.

    E ES

    HH

    E

    a

    b

    c

    d

    E+

    S

    SEAr

    HS

    σ-complexcation intermediate

    addition adduct

    H+E

    π-complex

    e

    E+

    SH

    HEf

    and/or

    g

    Figure 1 The typical mechanism of aromatic substitution reaction

  • 77

    In order for a substitution to occur, a “σ-complex” must be formed.

    The term σ-complex is used to describe an intermediate in which the

    carbon at the site of substitution is bonded to both the electrophile and the

    hydrogen is displaced. As the term implies, a σ bond is formed at the site

    of substitution. The intermediate is a cyclohexadienyl cation. Its

    fundamental structural characteristics can be described in simple MO

    terms. The σ-complex is four-π-electron delocalized system that is

    electronically equivalent to a pentadienyl cation. There is no longer cyclic

    conjugation. The LUMO has nodes at C-2 and C-4 of the pentadienyl

    structure, and these positions correspond to the position meta to site of

    substitution on the aromatic ring. As result, the positive charge of the

    cation is located at the positions ortho and para to the site of substitution.

    In electrophilic substitution, the formation of the σ complex is

    generally the rate-determining step, with the aromatization occurring

    much faster than the addition of the nucleophile to the σ complex

    carbocation, but there are exceptions. Some authors indicate that

    nucleophile addition proceeds faster than deprotonation,2 but the inability

    to isolate the intermediate adducts - due to their rapid rearomatization or

    further reaction to multi-addition products - forces investigators to draw

    conclusions regarding intermediate identity based solely on structural

    information obtained from the products as shown in Scheme 1, 2 and 3.

  • 78

    NF

    H

    ClCH2Cl

    -OAcN

    CH2Cl2

    NCH2Cl2

    NF

    H

    C lNF

    H

    OAc

    AcOF

    -HF

    N Cl N OAc+ +

    F2, AcOH

    Scheme 1. The substitution of pyridine via addition intermediate.2a

    NCH3

    NCS

    NCH3

    Cl N

    O

    O

    -HCl

    NCH3

    N

    O

    O74%

    NaHCO3,CHCl3

    Scheme 2. The substitution of N-methylpyrrole via addition intermediate.2b

    CH3

    C l

    CH3

    C l HH

    Cl+

    NH2-

    CH3

    H

    -H+

    CH3

    C l H

    NH2H

    -HCl

    CH3

    NH2

    C lH2N AlC l3

    NH2C l, AlC l3NH2C l + AlC l3

    C l+[H2NAlC l3]-

    +

    67% 13%

    Scheme 3. The chlorination and amination of toluene.2c

  • 79

    In cases where isolated adducts have been identified, the

    intermediates sometimes imply significant mechanistic differences when

    compared to the majority of known electrophilic aromatic substitutions.

    One example of this is the series of adducts identified in the nitration of

    furan (Scheme 4), whose mode of decomposition differs greatly from that

    commonly seen in 6-membered systems.3

    O

    HNO3--AcOH

    OO2NAcO-

    OO2N OAc

    46%

    Scheme 4. The electrophilic aromatic addition of furan.

    During preparation of 5,7-dibromo-8-methoxyquinaldine4 as a key

    intermediate in the synt