You are on page 1of 97

Analgesics and Antipyretics 1

Analgesics and Antipyretics


Elmar Friderichs, Grünenthal GmbH, Center of Research, Aachen, Germany
Thomas Christoph, Grünenthal GmbH, Center of Research, Aachen, Germany
Helmut Buschmann, Laboratorios Dr. Esteve S.A., Barcelona, España

1. Introduction . . . . . . . . . . . . . 1 3. Centrally Acting Analgesics . . 42


2. Nonsteroidal Anti-inflammatory 3.1. Opioids . . . . . . . . . . . . . . . . 42
Drugs (NSAIDs, COX 3.1.1. Opioids in Clinical Use . . . . . . 49
Inhibitors) . . . . . . . . . . . . . . 3 3.1.1.1. Morphine and Morphinane
2.1. Salicylates . . . . . . . . . . . . . . 7 Derivatives . . . . . . . . . . . . . . 49
2.2. para-Aminophenol Derivatives 8 3.1.1.2. Piperidine Derivatives . . . . . . . 56
2.3. Anthranilates . . . . . . . . . . . . 10 3.1.1.2.1. Meperidine and Congeners . . . . 56
2.4. Arylacetic Acids . . . . . . . . . . 12 3.1.1.3. Fentanyl and Congeners . . . . . . 58
2.5. Arylpropionic Acids . . . . . . . 19 3.1.1.4. Methadone and Congeners . . . . 60
2.6. Pyrazolinone Derivatives . . . . 27 3.1.2. Other Structures . . . . . . . . . . . 64
2.7. Acidic Enolic Compounds . . . . 28 3.1.2.1. Mixed Opioid Agonist – Antago-
2.7.1. Pyrazolidine-3,5-diones . . . . . . 28 nists and Partial Agonists . . . . . 66
2.7.2. Arylsulfonamides (Oxicames) . . 30 3.1.2.2. Opioid Antagonists . . . . . . . . 72
2.8. Other Structures . . . . . . . . . . 32 3.2. Antineuropathic Analgesics and
2.9. COX-2 Inhibitors . . . . . . . . . 33 Other Non-opioid Compounds . 74
2.9.1. Nonselective COX-2 Inhibitors . 33 3.2.1. Neuropathic Pain Treatment . . . 74
2.9.2. Selective COX-2 Inhibitors 3.2.2. Individual Compounds . . . . . . . 76
(Coxibs) . . . . . . . . . . . . . . . . 35 4. References . . . . . . . . . . . . . . 85

1. Introduction supposed additional mechanisms are widely un-


known.
Compounds that are used for the treatment The group of centrally acting analgesics is
of pain, inflammation, and fever are classified dominated by the opioid compounds. They act
into two main groups according to their mode mainly within the central nervous system, but
of action: nonsteroidal anti-inflammatory drugs more recent investigations indicate that an ad-
(NSAIDs) and so-called centrally acting anal- ditional peripheral action component may ex-
gesics. ist. The endogenous opioid system is the most
Nonsteroidal anti-inflammatory drugs are important pain control system within the body
compounds with a predominantly peripheral and opioids are powerful tools for the treatment
mechanism of action. Besides inhibition of pain of severe pain situations. Central neurotransmit-
they exert a more or less pronounced anti- ters like noradrenaline and serotonin are like-
inflammatory and fever-reducing effect. In the wise involved in pain inhibition. Directly acting
body key peripheral mediators of pain, in- compounds like, e.g., the α2 -adrenergic agonist
flammation, and fever are prostaglandins (→ clonidine as well as indirectly acting inhibitors
Prostaglandins). NSAIDs and related “periph- of noradrenaline and serotonin reuptake (mainly
eral” or “weak analgesics” act via inhibition of used as antidepressants) have a definite value as
cyclooxygenase. Cyclooxygenase is the key en- primary analgesics or as co-analgesics in com-
zyme of prostaglandin synthesis and its inhibi- bination with opioids.
tion may be the most important, but not the ex- Acute pain is an alarm signal to alert the or-
clusive mechanism of action of NSAIDs. The ganism to noxious tissue damage, irritation, or
injury. Ongoing, persistent or chronic pain may

c 2007 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim


10.1002/14356007.a02 269.pub2
2 Analgesics and Antipyretics

lose this function and may become a mere bur- Inhibitory control of the pain process can take
den to the patient. Pain does not only indicate place in the periphery at the level of the nocicep-
physical suffering, but involves a strong men- tors or the primary afferents. These are the re-
tal and emotional component. According to the gions where local anesthetics and NSAIDs act.
definition of the International Association for the Central pain inhibition can take place at the level
Study of Pain “it is an unpleasant sensory and of the spinal cord, the thalamus, cortex, and the
emotional experience associated with actual or limbic system. The pain transmission at the level
potential tissue damage, or described in terms of of the spinal cord is under inhibitory control of
such damage”. descending fibers, which originate in different
The nervous system contains an afferent fiber parts of brain stem, mid brain, and cortex. They
system that is specialized for registration, trans- contain opioid synapses as well as noradrener-
mission, and processing of nociceptive infor- gic and serotoninergic synapses and represent
mation. In the periphery small unmyelinated the descending spinal pain inhibitory system.
nerve endings, called nociceptors, are activated Inhibitory opioid synapses are not only found
by mechanical, thermal, or chemical stimuli. in the spinal cord but are strategically located
One type, the mechano-heat nociceptor is spe- along the whole pain pathway. Opioid receptors
cialized for thermal and mechanical stimulation, are found in the cortex, limbic system, and for-
whereas the so-called polymodal receptor is less matio reticularis and this explains why opioids
specific and responds to all kinds of mechan- not only inhibit pain perception but also reduce
ical, thermal, or chemical irritation. Mechano- the emotional component of pain suffering and
heat nociceptors have a high stimulation thresh- mitigate the vegetative pain symptoms.
old and respond only to strong stimuli. They According to the clinical situation, pain can
are linked to small myelinated and rapidly con- be classified as acute and chronic pain. Acute
ducting Aδ fibers which produce a sharp and pain is induced by trauma, tissue damage, or
well located pain sensation. The sharp pain stim- disease and has a well defined localization and
ulus activates withdrawal reflexes and has to time course. Acute pain normally responds well
protect the body against further damage. The to NSAIDs and opioid analgesics. Chronic pain
polymodal receptors respond to weaker stim- is more complex in nature and often contains an
uli including endogenous compounds like his- inflammatory component. The association with
tamine, bradykinin, and prostaglandins. These trauma and lesion can be lost and then pain
receptors are mediators of pain and inflamma- no longer fulfills its alarming function. Chronic
tion and are released by tissue damage. The im- pain is often a heavy burden to the patient and
pulses of polymodal nociceptors are transmitted induces physical, psychological, and social dete-
more slowly via thicker, unmyelinated C-fibers rioration. Chronic pain treatment is an important
and provoke a dull, aching, and less precisely medical challenge.
localized pain sensation. The nerve fibers origi- Physiologically, pain can be differentiated
nating from nociceptors are named primary af- into nociceptive and neuropathic pain. Nocicep-
ferent fibers. They have their cell body in the tive pain results from activation of nociceptors
dorsal root ganglia and project to the dorsal root by acute or chronic stimulation and is not associ-
of the spinal cord where they form synapses with ated with damage of nerves. Neuropathic or neu-
fibers of the ascending spino-thalamic pathway. rogenic pain in contrast is the result of damage
Most of the spino-thalamic pain fibers pass the or dysfunction of the peripheral nerves or of the
thalamus and project to the limbic system and central parts of the pain processing system. Ex-
the cortex. The limbic system is responsible for amples of neuropathic pain of peripheral origin
the emotional component of pain whereas in the are deafferentiation pain (stump pain), diabetic
cortex realization and localization of the origin neuropathy, post herpetic neuralgia, trigeminal
of pain takes place. The spino-thalamic fibers neuralgia, and sympathetically maintained pain
form connections with the formatio reticularis like the complex regional pain syndrome. Cen-
of the brain stem, inducing the various vegeta- tral neuropathic pain may result from lesions of
tive reactions associated with the pain percep- the spinal cord or brain lesions following stroke.
tion such as sweating, palpitation, nausea, and In chronic cancer pain, nociceptive pain is of-
blood pressure increase. ten associated with a neuropathic component in-
Analgesics and Antipyretics 3

duced by neoplastic nerve infiltration or com- inflammatory drug and is used in almost every
pression of the nerve by tumor growth. household for the treatment of mild to moder-
Cancer pain is one of the most important ate pain states and fever. In 1971, it was shown
forms of chronic severe pain and guidelines for that the analgesic action of nonsteroidal anti-
the treatment of cancer pain were published by inflammatory drugs is due to the inhibition of
the WHO in 1986 (Fig. 1). the enzymatic production of prostaglandins. Cy-
clooxygenase (COX) is the key enzyme in the
conversion of arachidonic acid derived from
lipids of the cell membrane to prostaglandins
and other eicosanoids (Fig. 2).

Figure 1. WHO-Guidelines for Cancer pain treatment


(“WHO-Ladder”)

Meanwhile these guidelines have become the


standard not only for treatment of chronic malig-
nant pain but also for benign chronic and acute
pain. The WHO proposal is often described as
“treatment by mouth, by the clock, and by the
ladder”. That means that for repeated applica- Figure 2. Simple scheme of the biosynthesis of
prostaglandins (PG), thromboxanes (TX), and leukotrienes
tion of analgesics the oral route should be used. from arachidonic acid consequent to cell injury (modified
The application interval should be regular and after [6])
selection of compounds should follow the in-
crease of potency and efficacy as indicated in the The eicosanoids are important mediators in
three-step analgesic ladder. The first step starts pain and inflammation leading to hyperalgesia
with the single use of a non-opioid analgesic. If by sensitization of nerve fibers and fever. Fur-
pain control is insufficient, a weak opioid may be thermore they fulfill important roles in the pro-
added. If this is not effective enough, the weak tection of the gastric mucosa, in platelet aggre-
opioid should be replaced by a strong opioid gation, and maintenance of normal kidney func-
and the non-opioid may be omitted. Each stage tion [11]. The diversity of physiological func-
of the treatment may be supplemented by the tions of the eicosanoids is reflected by a variety
use of co-analgesics and other pharmacological of different receptors. Five main receptor types
and non-pharmacological treatments to improve have been described, designated DP, FP, IP, TP,
pain control and to reduce side effects. and EP, which show the greatest apparent affinity
for PGD, PGF, PGI2 , TXA2 , and PGE, respec-
tively [12]. PGE2 , the eicosanoid which plays a
2. Nonsteroidal Anti-inflammatory key role in pain perception, exerts this and other
Drugs (NSAIDs, COX Inhibitors) functions via the four subtypes of the EP recep-
tors, EP1 – 4.
In 1899 acetylsalicylic acid, the first non- COX exists in two different isoforms, COX-1
steroidal anti-inflammatory drug was regis- and COX-2, characterized by different expres-
tered under the name aspirin. Acetylsalicylic sion patterns. Both forms are encoded by two
acid is still the best-known nonsteroidal anti- genes which have been cloned and sequenced
4 Analgesics and Antipyretics

[13–16]. The cloning of the two isoforms led → Prostaglandins, Chap. 3). As expected, the
to further characterization of the enzymes by gastrointestinal side effects of these drugs were
means of a whole range of molecular biology shown to be clearly lower than those of clas-
tools from knock-out mice [17–19] to protein sical nonsteroidal anti-inflammatory drugs like
structures [20, 21]. naproxen.
COX-1 is constitutively expressed in many The ongoing research on the COX isoforms
cells of the body and responsible mainly for the revealed that the strict differentiation of COX-
production of eicosanoids serving normal physi- 1 as the constitutive enzyme and source of
ological functions. One important physiological physiological (good) prostaglandins and COX-
role is the protection of the gastric mucosa. Pain 2 as the inflammation- and pain-mediated in-
relief by inhibition of COX-1 is therefore often duced form of the enzyme, producing the “bad”
accompanied with gastrointestinal side effects. prostaglandins is no longer valid. It was shown
COX-2 expression is induced during inflam- that COX-1 expression is also subject to regula-
mation and is thought to be responsible for the tory processes and can be increased in inflam-
production of eicosanoids in inflammatory con- matory conditions [25]. COX-2 expression on
ditions related to fever and pain. Furthermore, the other hand is increased in the gastric mucosa
COX-2 is expressed in the central nervous sys- by inflammatory stimuli and Helicobacter pylori
tem and might play a direct role in central pain infection [26, 27]. Finally, analysis of COX-1
processing. and COX-2 knock-out mice indicated that both
Many nonsteroidal anti-inflammatory drugs isoforms can contribute to an inflammatory re-
of different chemical structures have been in- sponse and have significant roles in the mainte-
troduced for the treatment of inflammatory and nance of physiological homeostasis and carcino-
painful conditions. Many years of clinical expe- genesis [28]. These data suggest a more com-
rience with these drugs have shown that there plex situation than the initial idea on the roles of
is no induction of tolerance and dependence COX-1 in normal and of COX-2 in inflammatory
and no respiratory depression as seen with opi- conditions, respectively.
oids. The major side effects of these compounds Nevertheless, the COX-2 inhibitors showed
with COX-1 specificity or balanced COX-1 and a clearly improved gastrointestinal side effect
COX-2 inhibition are damage of the gastric mu- profile and up to the year 2000 the newly de-
cosa, prolongation of bleeding time, and renal veloped coxibs seemed to displace the classical
failure. The discovery of the inducible isoform COX-1 selective NSAIDs from the market. The
led to the identification of drugs that show a situation totally changed when the results of on-
stronger inhibition of COX-2 compared to COX- going clinical studies and case reports revealed
1 (Table 1). that COX-2 inhibitors seem to increase the risk
The characteristic differences in expression of adverse thromboembolic events. The case re-
of the two COX isoforms suggest a potential for ports came from patients with connective tissue
new drugs addressing inflammatory and painful diseases who developed arterial thrombosis after
conditions specifically via inhibition of COX-2. starting celecoxib treatment [29]. An increased
The analgesic and anti-inflammatory potential number of cardiovascular events as compared to
of selective COX-2 inhibitors should come with- naproxen were seen in a large-scale randomized
out the well known gastrointestinal side effects controlled trial [30], which investigated the ef-
of classical NSAIDs targeting COX-1 alone or ficacy and tolerability of rofecoxib (the VIGOR
in combination with COX-2. trial).
The first generation of COX-2 inhibitors like The concern about the risk associated with
meloxicam still shows inhibition of COX-1 in the COX-2 inhibitors increased rapidly and in
physiological concentrations and hence, still September 2004 rofecoxib was withdrawn from
generates gastrointestinal side effects of COX- the market because a trial, designed to test the
1 inhibition although to a lower degree. The hypothesis that COX-2 inhibitors would pre-
second generation of COX-2 inhibitors reached vent recurrent colonic polyps, indicated an in-
the market in 1999 with celecoxib and rofe- creased cardiovascular toxicity [31]. A similar
coxib and shows a more than 100-fold selec- trial with celecoxib, performed at the National
tivity for COX-2 compared to COX-1 (see also Cancer Institute was stopped when an indepen-
Analgesics and Antipyretics 5
Table 1. COX-2 specific inhibitors
Group Compound COX-2 selectivity Reference
First generation (< 100-fold)
etodolac 10 [22]
meloxicam 10 [22, 23]
nimesulide 5 – 100 [22, 23]
Second generation (100 – 1000-fold)
celecoxib 375 [24]
rofecoxib 800 [561]

dent panel of cardiovascular experts reviewed ciples such as inhibition of 5-lipoxygenase (5-
the data and also found a greater incidence of car- LO) or release of nitric oxide (NO).
diovascular events among patients treated with
celecoxib [32]. Negative data on cardiovascu- Dual COX and 5-LO Inhibitors. This
lar tolerability were also seen with valdecoxib new class of compounds is reported to have
and its intravenous prodrug, parecoxib, given COX-2 and 5-lipoxygenase inhibiting proper-
for pain treatment to patients recovering from ties [35]. The compounds were effective in sev-
coronary-artery bypass surgery [33]. In the be- eral animal models of arthritis and were devoid
ginning, the mechanism of the cardiovascular of ulcerogenic properties. One of these com-
toxicity of the COX-2 inhibitors was unclear, but pounds, S-2474, ((E)-5-(3,5-di-tert-butyl-4-hy-
it emerged that it was a group effect of the coxibs droxybenzylidene)-2-ethyl-1,2-isothiazolidine-
and the most likely mechanism is a reduced pro- 1,1-dioxide), which has a γ-sultam skeleton, ad-
duction of the antithrombotic eicosanoid prosta- ditionally inhibits the production of interleukin-
cyclin without changing the production of the 2 (IL-2) in in vitro assays. S-2474 was selected
prothrombotic compound thromboxane [34]. as development candidate and is now under
Besides the cardiovascular intolerability, the clinical investigation [36].
COX-2 inhibitor valdecoxib showed an in- ML-3000 ((2,2-dimethyl-6-(4-chlorophe-
creased risk of serious and potentially fatal skin nyl)-7-phenyl-2,3-dihydro-1H-pyrrolizine-5-
reactions which was the reason for the suspen- yl)-acetic acid) is a nonoxidant dual cy-
sion of marketing autorization. The remaining clooxygenase and 5-lipoxygenase inhibitor and
COX-2 inhibitors, etoricoxib and lumiracoxib, has been compared with indometacin in a num-
although having in principle a similar cardiovas- ber of experimental models of inflammation
cular and skin tolerability risk, seem to be better and pain [37]. Corresponding to animal data
tolerated and are still on the market, but doctors the compound showed a wide range of ac-
are advised to use COX-2 inhibitors only after tivities in Phase II clinical studies, including
careful consideration, and to treat their patients anti-inflammatory, analgesic, antiplatelet and
with the shortest course and the lowest possible antiasthmatic properties [38, 39].
dose of these compounds.
The unexpected tolerability problems with NO Releasing COX Inhibitors. Nitric
the COX-2 inhibitors have strongly reduced the oxide (NO) releasing non-steroidal anti-
enthusiasm originally evoked by these com- inflammatory drugs are a new and promising
pounds and have re-established the therapeutic goup of analgesic and anti-inflammatory com-
reputation of the COX-1 inhibitors and of com- pounds. NO release is found as a mechanistically
pounds with a balanced COX-1 and COX-2 in- not yet resolved property of a few new COX-
hibition, although the gastrointestinal side ef- 1 inhibitors such as amtolmetin [40] or it can
fects remain a serious problem. Therefore there be induced by adding a NO releasing moiety
is increased need for new anti-inflammatory (e.g., nitroxybutyl) to existing COX-inhibitors
principles with better gastrointestinal tolerabil- [41]. The most advanced compounds are NO-
ity and research activities have been focussed naproxen and NO-flurbiprofen (NCX-2216).
on new compounds combining COX-inhibition Preclinical and clinical studies confirm that
with other analgesic and anti-inflammatory prin- both NO-release and COX-1 or COX-2 inhibi-
6 Analgesics and Antipyretics

tion take place in vivo, causing less adverse ef- mimics in vivo conditions like plasma binding
fects on the gastrointestinal tract than conven- [45]. It is commonly accepted that reasonable
tional NSAIDs and coxibs. In addition, the com- variations in COX inhibition is found in differ-
pounds reduce systemic blood pressure. Possi- ent laboratories. Therefore, whenever possible,
ble indications for NO-flurbiprofen are, besides data of several compounds, generated within the
pain inhibition and anti-inflammation, urinary same test situation, should be compared to each
incontinence, Alzheimer’s disease, osteoporosis other.
and Paget’s disease [42]. A topical formulation Using this classification based on isoenzyme
is under development for the treatment of der- selectivity, there is besides low-dose aspirin no
matological disorders such as contact urticaria. COX-1 selective inhibitor on the market. In-
A further group of NO-donating drugs (e.g., stead, most of the classical NSAIDs belong to
NCX-4016) has been obtained by coupling the group of nonselective COX inhibitors.
NO to aspirin. These compounds retain the
analgesic, anti-inflammatory and antithrombotic • Selective COX-1 inhibitors (low-dose as-
properties of aspirin but show an increased pirin)
gastrointestinal tolerability [43]. Animal stud- • Nonselective COX inhibitors (e.g., high-dose
ies have shown that NO-aspirins and the NO- aspirin, indometacin, and other NSAIDs)
donating NSAIDs maintain gastric mucosal • Preferential COX-2 inhibitors (e.g., meloxi-
blood flow and reduce leucocyte-endothelial cell cam)
adherence. • Highly selective COX-2 inhibitors (e.g., cele-
Up to 2006, no NO-COX inhibitor compound coxib and other second- and third-generation
has yet reached the market. COX-2 inhibitors, now called coxibs)
Classification According to Drug – Protein
Aspirin as an Antithrombotic Drug. As- Interactions. Another classification is based on
pirin, the archetype of the COX inhibitors, is still the mode of interaction between the inhibitor
one of the most interesting compounds of this drug and the enzyme [46].
group and has reached additional importance in
the last decades due to its inhibition of platelet • Irreversible inhibitors of COX-1 (aspirin)
aggregation. It is used in low dose [44] in the and COX-2 (APHS). Aspirin and APHS [47]
treatment of acute and the prevention of chronic acetylate the amino acid serine in the cat-
myocardial infarction and stroke (→ Cardiovas- alytic center of the enzyme to a stable ester
cular Drugs). derivative so that the endogenic arachidonic
acid is prevented from interaction with the
Classification of COX Inhibitors. COX in- enzyme.
hibitors, as it is done here, can be classified ac- • Reversible, competitive inhibitors of COX-
cording to their chemical structure. 1 and COX-2. Inhibitors such as ibuprofen,
Classification According to Inhibition Speci- piroxicam, or mefenamic acid block the en-
ficity. A more functional classification is related zyme by competing with arachidonic acid for
to the specificity for inhibition of the COX-1 binding at the catalytic center.
or COX-2 isoforms of the enzyme. The inhi- • Slow, time-dependent, reversible inhibitors
bition of the COX isoenzyme is strongly de- of COX-1 and COX-2. Compounds such as
pendent on the test system used. Inhibition of indometacin and flurbiprofen act by ionic in-
COX can be quantified in recombinant or natu- teraction between the acidic carboxyl group
ral enzyme preparations, cellular systems, iso- of the inhibitor and the basic arginine residue
lated human cell populations, such as platelets of the enzyme. This seems to influence the
(COX-1) and white blood cells (COX-2), or in helix D of the protein followed by a signifi-
ex vivo stimulated whole blood samples. The cant loss of flexibility of the enzyme protein.
more physiological the situation is, the smaller • Slow, time-dependent, irreversible inhibitors
is the selectivity of most of the COX-2 selec- of COX-2. The coxibs such as celecoxib, rofe-
tive inhibitors. The standard test for comparison coxib and lumiracoxib induce a strong, time-
is considered to be a whole blood assay which dependent inhibition of COX-2, but show
only a minor competitive inhibition of COX-
Analgesics and Antipyretics 7

1, which is of no relevance compared to the therapy (75–100 mg/d). It is also available in


COX-2 inhibition. rectal and topical formulations and as soluble
lysine derivative for intravenous or intramuscu-
COX Inhibitors in Clinical Use lar application. Acetylsalicylic acid is often used
in multi-drug preparations. The main side ef-
fects are gastrointestinal disorders. The use in
2.1. Salicylates children is limited due to the risk of Reye’s syn-
drome [53]. The lithium, magnesium, calcium,
Acetylsalicylic acid [50-78-2], aspirin, 2- and aluminum salts of acetylsalicylic acid are
acetoxybenzoic acid, C9 H8 O4 , M r 180.04, mp used in some special preparations.
135 ◦ C (for properties and synthesis, see → Trade names: Aspirine (France), Aspirin,
Salicylic Acid). Aspisol (Germany), Aspro (UK), Alka Seltzer
Synthesis [48, 49]: (USA).

Diflunisal [22494-42-4], 2 4 -di-


fluoro-4-hydroxybiphenyl-3-carboxylic acid,
C13 H8 F2 O3 , M r 250.20, mp 210–211 ◦ C (also
reported as 212–213 ◦ C).
Synthesis [48, 54–58, 505]: The diazotation
of 2,4-difluoroaniline with isoamyl nitrite and
Clinical use: Acetylsalicylic acid is the pro- condensation with anisole gives 4-(2,4-difluoro-
totype of a nonsteroidal anti-inflammatory drug phenyl)anisole, which is hydrolyzed with HI in
and is used in a large number of inflammatory refluxing acetic acid yielding 4-(2,4-difluoro-
and pain indications including musculoskeletal, phenyl)phenol. Finally this compound is carbox-
soft-tissue and joint disorders, headache, dys- ylated with K2 CO3 and CO2 at 175 ◦ C and 90
menorrhoea, and fever [50]. In addition acetyl- bar, followed by an acidification.
salicylic acid is used as an antiplatelet drug
for the prevention of myocardial infarction and
stroke [44] and in combination with thrombolyt-
ics in the acute treatment of myocardial infarc-
tion.
Depending on the assay system, acetylsali-
cylic acid shows a balanced inhibition of COX-1
and COX-2 [23] or a selectivity towards COX-1
[22]. In addition to COX inhibition acetylsal-
icylic acid modulates the activities of several
cellular kinases [51], which may contribute to
its anti-inflammatory effects.
The inhibition of COX-1 and COX-2 by
acetylsalicylic acid does not follow a competi-
tive mechanism like with other nonsteroidal anti-
inflammatory drugs but rather is due to a co-
valent enzyme inhibition via acetylation. After
absorption, acetylsalicylic acid is hydrolyzed to
salicylate which itself still shows some COX in-
hibition activity. Both compounds are bound to
plasma proteins to 80–90 %. The plasma elim-
ination half-life is about 15 min for acetylsal-
icylic acid and between 3 and 22 h for salicy-
late, depending on the dose [52]. Acetylsalicylic
acid is usually administered orally (0.5–8 g/d)
for pain and inflammation and for antiplatelet
8 Analgesics and Antipyretics

Clinical use: Diflunisal [60] is a nonsteroidal of mild pain conditions including musculoskele-
anti-inflammatory drug used in the treatment tal, soft-tissue, and joint disorders. It is now
of mild to moderate pain including osteoarthri- mostly substituted by newer COX inhibitors.
tis, rheumatoid arthritis, and primary dysmenor- Trade names: Percutalgine (France), Intral-
rhoea. It is used as base or lysine- or arginine-salt gin (UK), Anabar (USA).
for oral or parenteral application.
Diflunisal is a weak inhibitor of both COX-1
and COX-2 [61]. 2.2. para-Aminophenol Derivatives
Peak plasma concentrations are reached
within 2 to 3 h after oral dosing. Diflunisal is Paracetamol [103-90-2], acetaminophen,
strongly bound to plasma protein (> 99 %), has N-(4-hydroxyphenyl)acetamide, C8 H9 NO2 , M r
a long elimination half-life (8–12 h) and non- 151.16, mp 168–169 ◦ C.
linear elimination kinetics. Hence, it is used with Synthesis:
an initial loading dose (1000 mg) and a lower
maintenance dose (500–1000 mg/d). a) classical route [48, 64]:
The main side effects are gastrointestinal dis-
turbances, headache and rash.
Trade names: Dolobis (France), Dolobid
(Italy, UK, USA).

Ethenzamide [938-73-8], 2-ethoxybenz-


amide, C9 H11 NO2 , M r 165.19, mp 132–134 ◦ C.
Synthesis: Salicylamide is ethylated with di-
ethyl sulfate [48, 62].

b) Hoechst-Celanese process:

Clinical use: Ethenzamide is a nonsteroidal


anti-inflammatory drug used mainly in combina-
tion with other ingredients for the treatment of
mild to moderate pain including musculoskele-
tal and joint disorders.
Trade names: Trancalgyl (France).

Salicylamide [65-45-2], 2-hydroxybenz-


amide, C7 H7 NO2 , M r 137.14, mp 140 ◦ C.
Synthesis [48, 63]:

Clinical use [65]: Paracetamol has analgesic


and antipyretic properties, but no relevant anti-
inflammatory action. It is used for the treat-
ment of various mild to moderate pain con-
ditions and to reduce fever. Despite a long
Clinical use: Salicylamide shows analgesic therapeutic use, the mechanism of action of
and antipyretic efficacy and was used in multi- paracetamol is unclear. The compound shows
drug combinations for the treatment of a variety some weak inhibition of COX-1 and COX-2,
Analgesics and Antipyretics 9

and there are speculations that a third COX nine, which increase glutathione conjugation of
isoenzyme, named COX-3, may be inhibited the metabolite, are used as antidote.
[66]. Recently, a new and unexpected mech- Paracetamol is not soluble in aqueous solu-
anism through which paracetamol could exert tions and cannot be given parenterally. A soluble
its analgesic effect was detected [67, 68]. Fol- glycine prodrug derivative has been developed
lowing deacetylation to its metabolite p-amino- for parenteral use (propacetamol, see below).
phenol, paracetamol is conjugated with arachi- Trade names: Benuron (Germany, Switzer-
donic acid to give a compound named AM404. land), Dafalgan (France) Tylenol (Austria,
The metabolite AM404 affects several important Canada, USA, Spain), Alvedon (Norway, Swe-
targets in the nervous system involved in pain. den, UK).
AM404 inhibits purified COX-1 and COX-2 and Combination with codeine: Co-Tylenol
prostaglandin formation in lipopolysaccharide- (USA).
stimulated macrophages. AM404 is also a po- Combination with tramadol: Zaldiar (Ger-
tent activator of the ion channel TRPV1 (tran- many, Austria, France, Spain, Switzerland), Tra-
sient receptor potential cation channel, former macet (UK), Ultracet (USA).
classified as the vanilloid receptor VR1 ) and has Paracetamol is also used in many generic for-
an effect on cannabinoid CB1 receptors. Both mulations.
TRPV1 and CB1 receptors are involved in pain
and in thermoregulatory pathways. Propacetamol [66532-85-2], 4-
(acetylamino)phenyl N,N-diethylglycinate,
C14 H2 ON2 O3 , M r 264.33, is a soluble glycine
prodrug derivative of paracetamol. It is used as
hydrochloride ([66532-86-3], C14 H2 ON2 O3 ·
HCl, M r 300.79), for intramuscular or intra-
venous application and is rapidly metabolized
to free paracetamol [70].

Trade name: Pro-Dafalgan (Belgium,


France), Pro-Efferalgan (Italy)

Phenidine [62-44-2], phenacetin, N-


(4-ethoxyphenyl)acetamide, C10 H13 NO2 , M r
179.22, mp 134–135 ◦ C.
Paracetamol is one of the most popular anal- Synthesis [48]:
gesics as single drug or in multi-ingredient
preparations, often in combination with NSAIDs
or weak opioids. It is used orally or rectally as
suppositories, the oral dose range is 500–1000
mg every 4–5 h up to 4 g daily. Side effects are
rare and may include hematological reactions,
leucopenia, agranulocytosis and other hypersen-
sitivity reactions. Paracetamol has a narrow ther-
apeutic dose range and overdosage induces se-
vere liver and renal damage [69] via accumu-
lation of a toxic metabolite, N-acetylbenzoqui-
noneimine (NABQI). Acetylcysteine or methio-
10 Analgesics and Antipyretics

Clinical use: Phenidine is a weak anal- Synthesis: 2-Chlorobenzoic acid is reacted


gesic, antipyretic compound without anti- with 3-trifluoromethylaniline in the presence of
inflammatory action. It has been used in com- copper and potassium carbonate [48, 74, 75].
bination with other compounds like aspirin, caf-
feine, or codeine, but due to hematological and
nephrotoxic side effects [71] has been with-
drawn from the market and substituted by the
less toxic paracetamol.

2.3. Anthranilates

Etofenamate [30544-47-9], 2-(2-hydroxy-


ethoxy)ethyl 2-(3-trifluoromethylphenylamino)
benzoate, C18 H18 F3 NO4 , M r 369.34, pale yel-
low viscous oil, thermolabile at 180 ◦ C, bp
(133.32 Pa) 130–135 ◦ C.
Clinical use: Flufenamate is a nonsteroidal
Synthesis: Esterification of the potassium salt
anti-inflammatory drug used for the treatment of
of flufenamic acid (see page 10) with 2-(2-
mild to moderate pain of musculoskeletal, joint
chloroethoxy)ethanol in dimethyl formamide as
or soft-tissue origin. It was marketed in a variety
solvent yields etofenamate [48, 72].
of topical formulations alone or in combination
with other ingredients.
Flufenamate is not recommended in patients
with acute porphyria and was associated with
acute proctocolitis [76].
In addition to COX inhibition flufenamate
like other fenamates modifies several ion chan-
nel functions, e.g., inhibition of nonselec-
tive cation conductance [77], calcium-activated
chloride channels [78], voltage-gated calcium
channels and potassium channels [79, 80] and
induces blocking of gap junctions [81]. The rel-
evance of these activities for the analgesic and
anti-inflammatory potential of fenamates is un-
Clinical use: Etofenamate [73] is a non- known.
steroidal anti-inflammatory drug which was
used mainly as a topical formulation (500–1300 Meclofenamic acid [644-62-2], meclofe-
mg/d) and by intramuscular injection (1 g/d) for namate, 2-[(2,6-dichloro-3-methylphenyl)ami-
the treatment of joint, musculoskeletal and soft- no]benzoic acid, C14 H11 Cl2 NO2 , M r 296.15,
tissue disorders. mp 257–259 ◦ C; monosodium salt monohy-
drate [6385-02-0], C14 H10 Cl2 NNaO2 · H2 O,
Flufenamic acid [530-78-9], flufe- M r 336.15, mp 289–291 ◦ C.
namate, 2-[3-(trifluoromethyl)phenyl]ami- Synthesis: Meclofenamic acid is obtained by
nobenzoic acid, C14 H10 F3 NO2 , M r 281.23, condensation of 2-bromobenzoic acid with 2,6-
mp 124-125 ◦ C (also reported as 134- dichloro-3-methylaniline using CuBr2 in dieth-
136 ◦ C); aluminum salt (3 : 1) [16449-54-0], yleneglycol dimethyl ether containing N-ethyl-
C42 H27 AlF9 N3 O6 , M r 867.66. morpholine, and heating at 145–155 ◦ C [48, 82–
84] .
Analgesics and Antipyretics 11

the gastrointestinal system and include diarrhea


[91].
Trade names: Ponstyl (France), Ponstan
(UK), Ponstel (USA).

Niflumic acid [4394-00-7], 2-(3-trifluoro-


methylphenylamino)nicotinic acid, 2-[[3-(tri-
fluoromethyl)phenyl]amino]-3-pyridinecarb-
oxylic acid, C13 H9 F3 N2 O2 , M r 282.22, mp
204 ◦ C.
Synthesis: Condensation of 2-chloronicotinic
acid with 3-trifluoromethylaniline or reaction of
Clinical use: Meclofenamate [85] is a non- 2-aminonicotinic acid with 1-bromo-3-trifluoro-
steroidal anti-inflammatory drug used for the methylbenzene yields niflumic acid [48, 92].
treatment of mild to moderate pain, muscu-
loskeletal, and joint disorders like rheumatoid
arthritis and osteoarthritis as well as dysmen-
orrhoea. Meclofenamate inhibits COX-1 and
COX-2 and modifies ion channels [86, 87] (see
page 10).
Meclofenamate is administered orally (300–
400 mg/d).
Trade names: Lenidolor (Italy), Meclomen
(Austria, Spain, Switzerland, USA).

Mefenamic acid [61-68-7], 2-


[(2,3-dimethylphenyl)amino]benzoic acid,
C15 H15 NO2 , M r 241.29, mp 230–231 ◦ C;
monosodium salt C15 H14 NNaO2 , M r 263.27.
Synthesis [48, 88]:
Clinical use: Niflumic acid [93] is a non-
steroidal anti-inflammatory drug used for the
treatment of inflammation and pain in muscu-
loskeletal and joint disorders like rheumatoid
arthritis as well as traumatic and postoperative
pain. Niflumic acid is used in oral, rectal, or top-
ical applications (up to 750 mg/d).
The morpholinoethyl ester morniflumate,
which is used in topical formulations, in-
hibits both cyclooxygenase and 5-lipoxygenase
which suggests an additional component of anti-
inflammatory activity [94].
Clinical use: Mefenamic acid is a non- Trade names: Niflurid, Niflugel (Belgium,
steroidal anti-inflammatory drug which is used France, Switzerland), Actol (Austria, Sweden).
for the treatment of mild to moderate pain con-
ditions, musculoskeletal and joint disorders like Tolfenamic acid [13710-19-5], 2-[(3-
rheumatoid arthritis and osteoarthritis, and dys- Chloro-2-methylphenyl)amino]benzoic acid,
menorrhoea. [89]. Mefenamic acid inhibits both N-(3-chloro-o-tolyl)anthranilic acid; N-
COX isoforms with some preference for COX-2 (2-methyl-3-chlorophenyl)anthranilic acid,
and modifies ion channels [90] (see page 10). C14 H12 ClNO2 , M r 261.70, mp 207.0–
Mefenamic acid is given orally (1500 mg/d 207.5 ◦ C.
maximal dose). The main side effects concern
12 Analgesics and Antipyretics

Synthesis: Tolfenamic acid is obtained by


condensation of 2-chlorobenzoic acid with 3-
chloro-2-methyl-phenylamine using CuBr2 in
diethylenglycol dimethyl ether [95, 96].

Clinical use: Tolfenamic acid is a non-


steroidal anti-inflammatory drug [97] used for
the treatment of rheumatoid arthritis [98], os-
teoarthritis, ankylosing spondylitis, trauma pain,
and dysmenorrhoea [99]. It is also used for the
prophylactic and acute treatment of migraine
[100]. The compound is further marketed for
veterinary use.
Trade name: Clotam (Denmark).

2.4. Arylacetic Acids

Acemetacin [53164-05-9], carboxymethyl


[1-(4-chlorobenzoyl)-5-methoxy-2-methyl-
1H-indol-3-yl]-acetate, C21 H18 ClNO6 , M r
415.82, mp 150–153 ◦ C (fine pale yellow crys-
tals).
Synthesis [48, 101] (see on top, right column).
In addition, the reaction product of in- Proglumetacin [57132-53-3], 1-(4-
domethacin (see page 14) with benzyl bromoac- Chlorobenzoyl)-5-methoxy-2-methyl-1H-
etate can be hydrogenated to acemetacin. indole-3-acetic acid 2-[4-[3[4-(benzoylamino)-
Clinical use: Acemetacin [102] is a non- 5-(dipropylamino)-1,5-dioxopentyl]oxy]pro-
steroidal anti-inflammatory drug acting directly pyl]-1-piperanzinyl]ethyl ester, rac-N-[2-
and via its major metabolite indomethacin. [1-(p-chlorobenzoyl)-5-methoxy-2-methyl-
Acemetacin is used in chronic joint pain as well 3-indolylacetoxy]ethyl]-N  -[3-(N-benzoyl-
as in postoperative pain. Oral dosing is recom- N  ,N  -di-n-propyl-dl-isoglutaminoyl)oxypro-
mended between 120 and 360 mg daily. pyl]piperazine, C46 H58 ClN5 O8 , Mr 844.43,
Trade names: Rantudil (Germany), Emflex dimaleate, 4-Benzamido-5-(dipropylamino)-
(UK). 5-oxopentanoic acid 3-[4-[2-[2-[1-(chloro-
Acemetacin is also used in several generic benzoyl)-5-methoxy-2-methyl-1 H-indol-3-
formulations. yl]acetoxy]ethyl]piperazin-1-yl]propyl es-
ter dimaleate [59209-40-4], C46 H58 ClN5 O8
2C4 H4 O4 , Mr 1076.58, mp 146–148◦ C.
Synthesis: The reaction of N-benzoyl-N  ,

N -di-n-propyl-dl-isoglutamin (proglumide)
Analgesics and Antipyretics 13

with 1-(3-chloropropyl)-4-(2-hydroxyethyl)-pi-
perazine obtained from 1-(2-hydroxyethyl)pi-
perazine and 1-bromo-3-chloropropane by
means of sodium methanolate in DMSO at
105◦ C gives N-[3-(N-benzoyl-N  , N  -di-n-
propyl-dl-isoglutaminyl)oxypropyl]-N  -(2-hy-
droxyethyl)piperazine, which is then conden-
sated with Indomethacin (1-(p-chlorobenzo-
yl)-5-methoxy-2-methylindole-3-acetic acid)
by means of dicyclohexylcabodiimide and
NaHCO3 in ethyl acetate [103, 104] (see on
top).
Proglumetacine is a prodrug of acemetacine
[105] and is used for the treatment of os-
teoarthritis, rheumatoid arthritis, gout, ankylos-
ing spondylitis, and pain [106].
Trade names: Protaxon (Gemany), Proxil
(Italy).

Bufexamac [2438-72-4], 2-(4-butoxy-


phenyl)-N-hydroxyacetamide, C12 H17 NO3 , M r
223.12, mp 153–155 ◦ C.
Synthesis [48, 107] (see right column).
14 Analgesics and Antipyretics

Clinical use: Bufexamac is a nonsteroidal clofenac is combined with misoprostol to reduce


anti-inflammatory drug used in topical formula- gastrointestinal side effects, which are the main
tions for mild skin disorders and as suppositories adverse events and with weak opioids (codeine)
(250–500 mg/d) for hemorrhoids. to increase the analgesic potential.
Trade names: Duradermal (Germany), Par- Trade names: Allvoran (Germany), Diclac
fenac (France, Germany). (Germany), Voltaren (Germany, Italy, USA),
Voltarène (France), Voltarol (UK).
Diclofenac [15307-86-5], [2-(2,6- Combination with misoprostol: Arthotec
dichlorophenylamino)phenyl]acetic acid, (Germany).
C14 H11 Cl2 NO2 , M r 296.15, mp 156–158 ◦ C; Combination with codeine: Combaren.
sodium salt [15307-79-6], C14 H10 Cl2 NNaO2 , Diclofenac is also used in many generic for-
M r 318.13, mp 283–285 ◦ C. mulations.
Synthesis [108]:
Indomethacin [53-86-1], [1-(4-chloro-
benzoyl)-5-methoxy-2-methyl-1H-indol-3-yl]-
acetic acid, C19 H16 ClNO4 , M r 357.79, mp 153–
154 ◦ C (crystals exhibiting polymorphism, mp
for another form is 162 ◦ C); sodium trihydrate
[74252-25-8], C19 H15 ClNNaO4 · 3H2 O, M r
433.82, pale yellow crystalline powder.
Synthesis [48, 110] (see next page).
Clinical use: Indomethacin is a nonsteroidal
anti-inflammatory drug commonly used for
the treatment of mild to moderate pain. In-
domethacin is used in acute and chronic pain
states like rheumatoid arthritis, osteoarthritis,
joint and soft-tissue pain, dental pain, postop-
erative pain and dysmenorrhoea. Indomethacin
is a COX-1 selective inhibitor with 10- to 60-
fold selectivity compared to COX-2 [22, 23]. It
is administered orally, rectally or topically (50–
150 mg/d, maximal daily dose 200 mg) as well
as ophthalmic solution.
The major side effects of indomethacin are
gastrointestinal irritations and inflammation and
central nervous system disturbances like depres-
sion, drowsiness, tinnitus, and convulsions. In-
Clinical use: Diclofenac [109] is a non- domethacin is absorbed after oral application
steroidal anti-inflammatory drug with balanced reaching peak plasma concentrations after 2 h.
COX-1 and COX-2 inhibition [22]. It is com- It is bound to about 99 % to plasma proteins and
monly used for a variety of inflammatory and shows a variable terminal half-life from 2.6 to
pain conditions such as musculoskeletal and 11.2 h in adults and up to 30 h in neonates.
joint disorders, periarticular disorders, soft- Indomethacin is available as sodium, meg-
tissue disorders, renal colic, acute gout, dysmen- lumine, or l-arginine salt or as prodrug
orrhoea, and postoperative pain. (proglumetacin maleate).
The plasma protein binding of diclofenac is Trade names: Elmetacin, Inflam, Wydora
greater than 99.5 % and the plasma elimination (Germany), Indocid (France, UK), Indocin
half-life is between 1 and 2 h. (USA), Indolgina (Italy), Indocolir eye drops
Diclofenac is used mainly as the sodium salt (Germany).
orally or parenterally (75–150 mg/d) and as oph- Prodrugs of indomethacin are acemetacin
thalmic solution. Topical formulations may con- (see Section page 12) and proglumetacin
tain the diethylammonium or epolamine salt. Di- maleate: Protaxon (Germany).
Analgesics and Antipyretics 15
a) Merck & Co. process [111]: b) Sumitomo process [112]:

Lonazolac [53808-88-1], [3-(4- is formed by adding calcium chloride to the free


chlorophenyl-1H-pyrazol-4-yl]acetic acid, acid [48, 113–115].
C17 H13 ClN2 O2 , M r 312.75, mp 150– Clinical use: Lonazolac [85] is a nonsteroidal
151 ◦ C; calcium salt (2 : 1) [75821-71-5], anti-inflammatory drug used for the treatment of
C34 H24 CaCl2 N4 O4 , M r 663.57, mp 270– acute inflammatory pain conditions of joint and
290 ◦ C (decomp.). soft-tissue disorders as well as posttraumatic and
Synthesis: The pyrazole-4-carbaldehyde syn- postoperative pain. Lonazolac is used as calcium
thesized according to the Vilsmeier synthesis is salt and is given orally (600 mg/d, initial dose up
reduced to the alcohol, which is chlorinated. The to 900 mg/d) or rectally (800 mg/d).
chloro derivative is reacted with sodium cyanide Trade names: Argun (Germany), Irritren
to give the nitrile, which is hydrolyzed to lona- (Austria, Belgium, Switzerland).
zolac. The calcium salt, slightly soluble in water,
16 Analgesics and Antipyretics

Sulindac [38194-50-2], (Z)-5-fluoro-


2-methyl-1-[4-(methylsulfinyl)phenyl]methy-
lene-1H-indene-3-acetic acid, C20 H17 FO3 S,
M r 356.41, mp 182–185 ◦ C (decomp.).
Synthesis [48, 116, 117] (see right column).
Clinical use: Sulindac [118] is a nonsteroidal Tolmetin [26171-23-3], [1-methyl-5-(4-
anti-inflammatory drug used in the treatment of methylbenzoyl)-1H-pyrrol-2-yl]acetic acid,
mild to moderate pain including musculoskele- C15 H15 NO3 , M r 257.28, mp 155–157 ◦ C;
tal and joint disorders like rheumatoid arthritis, sodium salt [35711-34-3], C15 H14 NNaO3 , M r
osteoarthritis, and gout. The main side effects are 279.27; sodium salt dihydrate [64490-92-2],
gastrointestinal disturbances and kidney stones C15 H14 NNaO3 · 2 H2 O, M r 315.30.
[119]. Sulindac induces no relevant COX inhibi- Synthesis [48, 122, 123] (see next page, left
tion whereas the active metabolite sulindac sul- column).
fide inhibits both isoenzymes with some COX-1 Clinical use: Tolmetin [124] is a nonsteroidal
preference [120], indicating that the pharmaco- anti-inflammatory drug used for the treatment of
logical activity of sulindac probably results from mild to moderate pain states in musculoskeletal,
its sulfide metabolite. Another metabolite, sulin- soft-tissue, and joint disorders like rheumatoid
dac sulfone, induces apoptosis in tumor cells and arthritis, osteoarthritis, and gout as well as juve-
sulindac is extensively studied for cancer treat- nile rheumatoid arthritis. Tolmetin inhibits both
ment [121]. Sulindac is administered orally or isoforms of cyclooxygenase with some prefer-
rectally (200–400 mg/d). ence for COX-1 [125]. Tolmetin is administered
Trade names: Arthrocine (France), Clinoril orally, rectally, or topically (600–1800 mg/d).
(Italy, UK, USA). The peak plasma concentrations are reached
within 30 to 60 min. Tolmetin shows a high
plasma protein binding of 99 % and a biphasic
plasma half-life of 1 to 2 and 5 h, respectively.
Analgesics and Antipyretics 17

Trade name: Tolectin (Austria, Switzerland,


UK, USA).

Amtolmetin guacil (AMG, ester pro-


drug of tolmetin) [87344-06-7], 2-[2-[1-
methyl-2-(4-methylbenzoyl)pyrrol-5-yl]acet-
amido]acetic acid 2-methoxyphenyl ester, 2-
[2-[1-methyl-5-(4-methylbenzoyl)pyrrol-2-yl]- Clinical use: Amtolmetin guacil (AMG) is
acetamido]acetic acid 2-methoxyphenyl ester, a non-steroidal anti-inflammatory drug devel-
N-[(1-methyl-5-p-toluoylpyrrol-2-yl)acetyl]- oped by Sigma-Tau for the treatment of pain
glycine 2-methoxyphenyl ester, 1-methyl-5-p- and inflammation. It acts similarly to tolmetin,
toluoylpyrrole-2-acetamidoacetic acid guaicyl and in preclinical trials displayed long-lasting
ester, C24 H24 N2 O5 , M r 420.46, mp not re- anti-inflammatory and analgesic activity. Am-
ported. tolmetin has a good gastrointestinal tolerability
Synthesis: The condensation of 1-methyl- in humans [127] and does not induce gastric ul-
5-(4-methylbenzoyl)pyrrole-2-acetic acid with cers in the rat [128]. The good gastrointestinal
glycine ethyl ester in the presence of car- tolerability and even a gastroprotective effect
bonyldiimidazole and triethylamine in tetra- was related to an increase of endogenous NO
hydrofuran gives the corresponding ethyl 2-[2- via stimulation of inducible NO synthase in the
(1-methyl-5-(4-methylbenzoyl)-1H-pyrrol-2- gastric mucosa [129].
yl)acetamido]acetate, which is hydrolyzed with Trade name: Eufans (Italy).
NaOH in tetrahydrofuran – water yielding 2-
[2-[1-methyl-5-(4-methylbenzoyl)pyrrol-2-yl]- Mofezolac [78967-07-4], 3,4-bis(4-meth-
acetamido]acetic acid. Finally, this compound is oxyphenyl)-5-isoxazoleacetic acid, 3,4-di-
esterified with 2-methoxyphenol (guaiacol) in (p-methoxyphenyl)isoxazole-5-acetic acid,
hot tetrahydrofuran with carbonyldiimidazole C19 H17 NO5 , M r 339.35, mp 147.5 ◦ C.
as catalyst [126].
18 Analgesics and Antipyretics

Synthesis:
a) The reaction of deoxyanisoin with hydroxyl-
amine in methanol gives the corresponding
1,2-bis(4-methoxyphenyl)ethanone oxime,
which is cyclized with ethyl acetate by means
of n-butyllithium in tetrahydrofuran yield-
ing 3,4-di(4-methoxyphenyl)-5-methylisox-
azole. Finally, this compound is condensed
with CO2 with n-butyllithium as catalyst
in tetrahydrofuran to yield mofezolac [130,
131]. The synthesis with ClCO2 C2 H5 in-
stead of CO2 is described in [132, 133].

Clinical use: Yoshitomi (now Mitsubishi


Pharma) has launched mofezolac, a COX-1 in-
hibitor [136] with preferential COX-1 inhibition
[137] for the treatment of arthritis-related pain
in Japan.
The compound is used in oral doses of 75 mg
for the treatment of periarthritis, lumbago, neck-
shoulder-arm syndrome and pain after surgery,
trauma, or dental extraction.
Trade name: Disopain (Japan).

Bromfenac [91714-94-2], 2-amino-3-


(4-bromobenzoyl)benzeneacetic acid, 2-[2-
amino-3-(4-bromobenzoyl)phenyl]acetic acid,
C15 H12 BrNO3 , M r 334.17, monosodium salt
[91714-93-1], C15 H11 BrNNaO3 , M r 356.15,
mp 284–286 ◦ C (dec.), monosodium salt sesqui-
hydrate [120638-55-3], C15 H11 BrNNaO3 3/2
H2 O, M r 766.35.
b) Alternatively the corresponding 1,2-bis(4- Synthesis: The cyclization of 2-amino-4’-
methoxyphenyl)ethanone oxime can be con- bromobenzophenone with ethyl 2-(methyl-
densed with (E)-methyl 3-methoxyacrylate thio)acetate with tert-butyl hypochlorite as
with the aid of potassium tert-butyrate in catalyst in dichloromethane at 70 ◦ C gives 7-
tert-butanol to yield (E)-methyl 4,5-bis(4- (4-bromobenzoyl)-3-(methylthio)-2,3-dihydro-
methoxyphenyl)-5-oxopent-3-enoate, which 1H-indol-2-one, which is desulfurized by treat-
is cyclized with hydroxyl amine. Subsequent ment with Raney nickel in THF yielding 7-(4-
treatment with oxygen in acetic acid yields bromobenzoyl)-2,3-dihydro-1H-indol-2-one.
mofezolac [132–135]. Finally, this compound is hydrolyzed with re-
fluxing 3 M aqueous NaOH and acidified with
concentrated HCl [138–141].
Analgesics and Antipyretics 19

Several additional ways for the synthesis of


nabumetone are described. A short, simple, and
economical process for large scale preparation
avoiding hazardous chemicals or tedious work
Clinical use: Bromfenac, used as the sodium up procedures has been published [149].
salt, is a non-steroidal anti-inflammatory drug Clinical use: Nabumetone [150] undergoes
acting via inhibition of COX-1 and COX-2. It rapid first-pass metabolism to the active metabo-
was launched in Japan in 2000 by Senju for lite 6-methoxy-2-naphthylacetic acid (6-MNA).
topical treatment of ocular inflammation [142] 6-MNA shows a balanced inhibition of both
and has been approved in the United States in COX-1 and COX-2 [22]. Nabumetone is used
2006 for the treatment of pain following cataract for the treatment of pain and inflammation asso-
surgery. It was used for the short-term treatment ciated with osteoarthritis and rheumatoid arthri-
of acute pain, but it was withdrawn for this in- tis. It shows a 99 % binding to plasma protein
dication in June 1998 because of several post- and an elimination half-life of up to 22 h with
marketing reports of severe hepatic failure [143– marked individual differences. Nabumetone is
145]. given orally (500–2000 mg/d).
The drug is rapidly absorbed and excreted. Trade names: Listran (Spain), Nabuser
The drug’s long duration of anti-inflammatory (Italy), Relafen (USA), Relifex (UK).
action despite its short half-life deserves further
investigation.
Trade name: Xibrom (Japan, USA). 2.5. Arylpropionic Acids

Nabumetone [42924-53-8], 4-(6-methoxy- Flurbiprofen [5104-49-4], 2-(3 -


naphthalen-2-yl)butan-2-one, C15 H16 O2 , M r fluorobiphenyl-4-yl)propionic acid, 2-
228.29, mp 80 ◦ C. fluoro-α-methyl[1,1 -biphenyl]-4-acetic acid,
Synthesis: Condensation of 6-methoxy-2- C15 H13 FO2 , M r 244.26, mp 110–111 ◦ C;
naphthaldehyde with acetone in an aque- sodium salt [56767-76-1], C15 H12 FNaO2 , M r
ous solution of NaOH gives 4-(6-methoxy-2- 266.25.
naphthyl)-3-buten-2-one, which is reduced with Synthesis [48, 151–154]:
H2 over Pd-C in ethyl acetate [48, 146–149].
20 Analgesics and Antipyretics

Clinical use: Flurbiprofen is a nonsteroidal


anti-inflammatory drug used for the treatment
of pain and inflammation associated with mus-
culoskeletal and joint disorders as well as neu-
ralgia, dysmenorrhoea, and postoperative pain.
Flurbiprofen is used as free acid or sodium
salt and given orally or rectally (150–200 mg/d,
max. 300 mg/d) and as ophthalmic solutions.
Peak plasma concentration appears 1 to 2 h after
oral application. The plasma protein binding is
about 99.5 % and the elimination half-life is in
the range of 3–5 h.
Use of flurbiprofen as antiplatelet agent after
myocardial infarction is discussed [155].
In addition to COX inhibition, flurbiprofen
induces weak inhibition of the transcription fac-
tors NFκ-B and AP-1 and inhibits cell prolifera-
tion, that is why its use in the treatment of cancer
has been investigated [156].
Trade names: Ansaid (USA), Antadys, Cebu-
tid (France), Dobedan (Germany), Froben (UK),
Ocufen (UK, USA). Ocuflur eye drops (Ger-
many)

Ibuprofen [15687-27-1], 2-(4-isobutyl-


phenyl)propionic acid, C18 H18 O2 , M r 206.28,
mp 75–77 ◦ C.
Synthesis [48, 157, 158] (see next page).
Several alternative processes are described in
the literature.
Clinical use: Ibuprofen [159] is a non-
steroidal anti-inflammatory drug, commonly
used for the treatment of mild to moderate
pain. It is used in conditions like rheumatoid
a) arthritis, osteoarthritis, joint and soft-tissue pain,
dental pain, postoperative pain, dysmenorrhoea,
and headache, including acute migraine attacks.
Ibuprofen is given by oral, rectal, or topical ad-
ministration (800–2400 mg/d) and with a lower
dose (40 mg kg−1 d−1 ) for the treatment of
fever in children.
Ibuprofen shows the typical side effects of
nonsteroidal anti-inflammatory drugs but seems
to be better tolerated than other nonsteroidal
anti-inflammatory drugs. This may be due to
the balanced inhibition of COX-1 and COX-2
as seen in cultured human cells [23].
Ibuprofen is absorbed within 1 to 2 h after
oral application, bound to plasma protein up to
99 % and shows a plasma half-life of about 2 h.
Ibuprofen is a racemate, the active enantiomer,
b) the S(+)-enantiomer, is available in some coun-
Analgesics and Antipyretics 21
a) Boots process (industrial process): b) Boots-Hoechst-Celanese process:

Combination with hydrocodone: Vicoprofen


(USA).
Combination with oxycodone: Combunox
(USA).
Ibuprofen is also marketed in many generic
formulations.

Ketoprofen [22071-15-4], 2-(3-benzo-


ylphenyl)propionic acid, 3-benzoyl-α-methyl-
benzeneacetic acid, C16 H14 O3 , M r 254.28,
mp 94 ◦ C; lysine salt (1 : 1) [57469-78-0],
C16 H14 O3 · C6 H14 N2 O2 , M r 400.47; sodium
salt [57495-14-4], C16 H13 NaO3 , M r 276.27.
Synthesis [48, 160] (see next page).
Clinical use: Ketoprofen [161, 162] is a non-
steroidal anti-inflammatory drug used for the
treatment of a variety of acute and chronic
pain and inflammatory conditions including
rheumatoid arthritis, osteoarthritis, ankylosing
spondylitis, postoperative pain, and dysmenor-
rhoea. It is administered orally, rectally, topi-
cally, or intramuscular (100–200 mg/d, maxi-
mal dose 300 mg/d) as sodium or lysine salt.
Ketoprofen is given as racemate. The pharma-
tries (dexibuprofen). Ibuprofen is given as free
cologically active isomer is mainly the S(+)-
base or a variety of salts, esters, and other deriva-
enantiomer, which is available in some coun-
tives. Low-dose ibuprofen is available without
tries as the trometamol (2-amino-2-(hydroxy-
prescription and has become one of the most
methyl)-1,3-propanediol) salt. As compared to
popular OTC analgesics.
the racemate, absorption of the S(+)-isomer is
Trade names: Actren, Anco (Germany),
said to be faster, leading to an earlier onset of
Brufen (France, Italy, UK), Ibuprox (Spain), Im-
action [163].
bun (Germany), Motrin (USA).
22 Analgesics and Antipyretics

a) First large-scale manufacturing process of


Syntex [167]:

b) Stereospecific Syntex process [168]:


The stereospecific Syntex process is an example
The peak plasma concentration after oral ad- using chiral technology to produce enantiomer-
ministration occurs within 2 h. Ketoprofen is ically pure naproxen:
bound to plasma protein up to 99 % and shows
a plasma elimination half-life of 1.5 to 4 h.
Trade names: Ketocid (UK), Profénid
(France), Orudis (Germany, Italy, USA).
S(+)-ketoprofen: Keral (UK).
Naproxen [22204-53-1], (+)-(S)-2-
(6-methoxynaphthalen-2-yl)propionic acid,
C14 H14 O3 , M r 230.26, mp 155.3 ◦ C (also
reported as 152–154 ◦ C), [α]D + 66◦ (c
= 1, CHCl3 ); sodium salt [26159-34-2],
C14 H13 NaO3 , M r 252.25, mp 244–246 ◦ C, [α]D
− 11◦ (c = 1, CHCl3 ).
Synthesis [48, 164–166]:
For racemic resolution of naproxen the use of
cinchonidine, N-alkyl-d-glucamine, dehydroa-
bietylamine or (S)-α-phenylethylamine is de-
scribed. For enzymatic cleavage of esters of
racemic naproxen, cloned esterases, which are
cheap and easy to produce, have been developed
(production in a 100 t/a process is planned). Sev-
eral other synthetic routes exist.
Analgesics and Antipyretics 23

The asymmetric hydrogenation of 2-(6-meth-


oxy-2-naphthyl)acrylic acid using ruthenium-
2,2 -bis(diphenylphosphino)-1,1 -binaphthyl
(BINAP) complexes yields also enantiomer-
ically pure naproxen.
Clinical use: Naproxen [169] is a non-
steroidal anti-inflammatory drug used for the
treatment of mild to moderate pain and inflam-
matory pain conditions like rheumatoid arthri-
tis, osteoarthritis, soft-tissue disorders, postop-
erative pain, and dysmenorrhoea. In contrast to
most other COX inhibitors, which are racemic
mixtures, naproxen has been developed and is
clinically used only as the (S)(+)-enantiomer.
Naproxen is given orally or rectally (up to 1250
mg/d). The major side effects are gastrointestinal
disturbances. To decrease gastrointestinal side
effects, fixed combinations wih the proton pump
inhibitor lansoprazole were developed.
Naproxen is available as free base, as sodium
salt, and in combination with misoprostol for the Zaltoprofen [7411-43-6], 2-(10-
reduction of the gastrointestinal side effects. For oxo-10,11-dihydrodibenzo[b,f ]thiepin-2-yl)-
topical use, morpholinyl- and methylpiperazi- propionic acid, α-methyl-10-oxo-10,11-di-
nylacyloxyalkyl prodrugs have been developed hydrodibenzo[b,f ]thiepin-2-acetic acid, 10,11-
[170]. dihydro-α-methyl-10-oxodibenzo[bf ]thiepin-
Trade names: Aleve (France, Germany), 2-acetic acid, C17 H14 O3 S, M r 298.36, mp
Anoprox (USA), Apranax (France, Germany), 131-133 ◦ C; (S)-enantiomer [89482-01-9], mp
Naprosyn (UK, USA), Proxen (Germany), Syn- 25
129.5-131 ◦ C), [α]D +32.4◦ (c = 1, chloroform).
flex (UK).
Synthesis: The cyclization of 5-(1-
Combination with lansoprazole: Prevacid,
cyanoethyl)-2-(phenylthio)phenylacetic acid in
NapraPAC (USA).
presence of polyphosphoric acid at 120 ◦ C gives
Naproxen is also marketed in several generic
2-(10-oxo-10,11-dihydrodibenzo[b,f ]thiepin-
formulations.
2-yl)propionitrile, which is then hydrolyzed
Tiaprofenic acid [33005-95-7], 2-(5-ben-
with KOH in refluxing ethanol – water to zalto-
zoylthiophen-2-yl)propionic acid, C14 H12 O3 S,
profen [174, 175].
M r 260.31, mp 96 ◦ C.
Synthesis [48, 171] (see right column, top).
Clinical use: Tiaprofenic acid [172] is a non-
steroidal anti-inflammatory drug used for the
treatment of mild to moderate pain states in mus-
culoskeletal, soft-tissue, and joint disorders as
well as for postoperative pain treatment. Tiapro-
fenic acid is given as oral or rectal administra-
tion (600 mg/d) and as intramuscular injection
of the trometamol (2-amino-2-(hydroxymeth-
yl)-1,3-propanediol) salt. The main side effects
are urinary tract symptoms like cystitis and blad-
der irritation [173].
Trade name: Surgam (France, Germany,
UK).
24 Analgesics and Antipyretics

Clinical use: Zaltoprofen is a non-steroidal yl)hydratropic acid, C13 H14 ClNO2 , M r 251.71,
anti-inflammatory drug originated by Nippon mp 98-100 ◦ C.
Chemiphar and jointly developed with Zeria. Synthesis:
It has been available on the market in Japan
a) Ethyl 4-nitrophenylacetate is methylated
since 1993 for the relief of pain and inflamma-
with NaH – CH3 I in dimethylformamide –
tion resulting from arthritis deformations, pe-
toluene to form ethyl 2-(4-nitrophenyl)-
riarthritis of the shoulder, neck-shoulder-arm
propionate, which is reduced with H2
syndrome, rheumatoid arthritis, lumbago, post-
over Pd/C in EtOH – H2 O to ethyl 2-(4-
surgery pain, trauma, and tooth extraction and
aminophenyl)propionate. This product is
used in oral doses of 80 mg [176]. According to
acetylated with acetic anhydride to yield
studies, the analgesic effects of zaltoprofen may
ethyl 2-(acetylaminophenyl)propionate. Al-
involve the inhibition of bradykinin-2 receptor-
ternatively, ethyl 4-aminophenylacetate can
mediated responses in primary afferent neurons
be acetylated with acetic anhydride to
[177, 178]. Although zaltoprofen is marketed as
ethyl 4-acetylaminophenylacetate followed
a racemate the anti-inflammatory activity resides
by methylation with CH3 I and sodium in
in the (S)-enantiomer.
liquid NH3 to give ethyl 2-(4-acetylami-
Trade name: Peon, Soleton (Japan).
nophenyl)propionate [179, 180]. Ethyl 2-(4-
acetylaminophenyl)propionate is chlorinated
Pirprofen [31793-07-4], rac-3-chloro-4-
with Cl2 in acetic acid to ethyl 2-(3-chloro-
(2,5-dihydro-1H-pyrrol-1-yl)-α-methylbenz-
4-acetylaminophenyl)propionate. This inter-
eneacetic acid, rac-3-chloro-4-(3-pyrrolin-1-
mediate is hydrolyzed with HCl in EtOH
Analgesics and Antipyretics 25

to give ethyl 2-(3-chloro-4-aminophenyl)- the treatment of post-operative pain, oncolog-


propionate hydrochloride. The last step is ical pain, episodic headache attacks, and acute
the reaction of the preceding intermedi- migraine [182]. There are reports on hepatic and
ate with cis-1,4-dichloro (or dibromo)-2- renal intolerability [183–186] which precluded
butene in dimethylformamide and Na2 CO3 the further use.
to ethyl 2-(3-chloro-4-(3-pyrrolinyl)phenyl)- Trade names: Rengasil (France, Belgium,
propionate, followed by hydrolysis with Netherlands, Chile), Seflenyl.
KOH in EtOH – H2 O (see previous page).
b) 2,4-Dichloronitrobenzene is reacted with Fenoprofen [31879-05-7], rac-α-meth-
sodium diethyl methylmalonate in dimeth- yl-3-phenoxybenzeneacetic acid, rac-m-
ylformamide to yield diethyl 2-(4-nitro-3- phenoxyhydratropic acid, α-(d,l)-2-(3-phen-
chlorophenyl)-2-methylmalonate, followed oxyphenyl)propionic acid, C15 H14 O3 , M r
by hydrolysis, decarboxylation and esteri- 242.27, viscous oil, bp0.11 (0.147 hPa) 168-
fication to form ethyl 2-(3-chloro-4-nitro- 171 ◦ C; calcium salt dihydrate, [53746-45-
phenyl)propionate. The latter is reduced 5], C30 H26 CaO6 . 2H2 O, M r 558.64, mp 68-
as in route (a) to ethyl 2-(3-chloro-4- 171 ◦ C.
aminophenyl)propionate hydrochloride. Al- Synthesis: The condensation of 3-hy-
ternatively, diethyl 2-(4-nitro-3-chlorophe- droxyacetophenone with bromobenzene yields
nyl)-2-methylmalonate can be hydrogenated 3-phenoxyacetophenone, which is reduced
first to diethyl 2-(4-amino-3-chlorophenyl)- with sodium borohydride to 1-(3-phenoxy-
2-methylmalonate, and then hydrolyzed, phenyl)ethanol. Bromination with phos-
decarboxylated and esterified again to form phorus(III)bromide yields 1-(1-bromoethyl)-
ethyl 2-(3-chloro-4-aminophenyl)propionate 3-phenoxybenzene. After reaction with sodium
hydrochloride [179, 180] (see below). cyanide the resulting 2-(3-phenoxyphenyl)pro-
panenitrile is hydrolyzed to fenoprofen [187,
Clinical use: Pirprofen is a nonsteroidal anti-
188]; for alternative syntheses see [189–191].
inflammatory drug [181] and can be used for
26 Analgesics and Antipyretics

by means of acetyl chloride. Methylation with


methyl iodide in the presence of n-butyllithium
yields the desired vedaprofen after the standard
work-up procedure [198].

Clinical use: Fenoprofen [192–194] is a non-


steroidal anti-inflammatory drug used in oral
doses of 300 mg as the calcium salt or in doses
of 200–400 mg as free base for the treatment of
moderate to severe pain and symptomatic treat-
ment of acute and chronic rheumatoid arthritis
and osteoarthritis. Enteric coated tablets of the
Ca salt were developed to reduce gastrointesti-
nal microbleeding [195]. The use of fenoprofen
was associated with renal impairment [196] and
allergic skin inflammation [197].
Trade names: Fepron (Italy), Nalfon (USA),
Nalgesic (France).
Clinical use: Vedaprofen is a nonsteroidal
Vedaprofen [71109-09-6], rac-4-cy- anti-inflammatory drug with antipyretic and
clohexyl-α-methyl-1-naphthaleneacetic acid, analgesic properties, based on inhibition of
rac-2-(4-cyclohexylnaphthalen-1-yl)propanoic prostaglandin synthesis. Both enantiomers con-
acid, C19 H22 O2 , M r 282.38, mp 150 ◦ C. tribute to the biological and therapeutic actions
Synthesis: The reaction of 1-cyclohexylnaph- of vedaprofen. Although vedaprofen was ini-
thalene and paraformaldehyde in the presence tially developed for human use, it is is now
of concentrated hydrochloric acid and H3 PO4 mainly used as a veterinary drug as oral gel [199]
at 85 ◦ C yields 1-chloromethyl-4-cyclohexyl- for the reduction of inflammation and pain asso-
naphthalene, which is transformed to (4-cy- ciated with musculo-skeletal disorders in dogs
clohexylnaphthalen-1-yl)-acetonitrile by means [200] and horses and parenterally as the meglu-
of sodium cyanide in water acetone mixture mine salt for the acute treatment of colic pain
as solvent. The hydrolysis of the nitrile with in horses [201]. In horses vedaprofen is admin-
potassium hydroxide yields the sodium salt of istered orally at an initial dose of 2 mg/kg, fol-
(4-cyclohexylnaphthalen-1-yl)-acetic acid. Af- lowed by 1 mg/kg every 12 h for up to 14 days.
ter treatment with hydrochloric acid the free Trade name: Quadrisol.
acid is converted to the corresponding (4-cyclo-
hexyl-naphthalen-1-yl)-acetic acid methyl ester
Analgesics and Antipyretics 27

Nepafenac [78281-72-8], 2-(2-amino-3- acetic acid in refluxing ethanol gives 1-(α-


benzoylphenyl)acetamide, 2-amino-3-benzoyl- methylphenethylidieneimino)indolin-2-one,
benzeneacetamide, C15 H14 N2 O2 , M r 254.29, which by reaction with refluxing ethanolic
mp 178.5-180 ◦ C. hydrogen chloride affords ethyl α-(2-methyl-
Synthesis: The reaction of (2-aminophenyl)- 3-phenylindol-7-yl)acetate. The ozonolysis of
phenylmethanone and thiocarbamic acid S- this intermediate in acetic acid yields ethyl 2-
methyl ester in the presence of 2,2-di- acetamido-3-benzoylphenyl acetate, which is
methylpropionyl chloride in methylene chloride cyclized by refluxing with HCl in acetic acid
at -65 ◦ C and subsequent treatment with triethyl- to give 7-benzoylindolin-2-one. Alternatively,
amine at the same temperature yields the cor- the hydrolysis of the ester ethyl α-(2-methyl-3-
responding condensation product 2-(2-amino- phenylindol-7-yl)acetate with KOH in refluxing
3-benzoyl-phenyl)-2-methylsulfanylacetamide. water affords the corresponding acid, which can
Finally, nepafenac is obtained by reduction with be ozonolyzed as before yielding 2-acetamido-
Raney nickel in tetrahydrofuran [202, 203]. 3-benzoylphenylacetic acid. This acid can be
cyclized to 7-benzoyl-1,3-dihydro-indol-2-one
by refluxing with HCl in acetic acid as before
[208–210].

2.6. Pyrazolinone Derivatives


Metamizol [50567-35-6], dipyrone[(1,5-
dimethyl-3-oxo-2-phenyl-2,3-dihydro-1H-
pyrazol-4-yl)methylamino]-methanesulfonic
acid, C13 H17 N3 O4 S, M r 311.36; sodium
salt [68-89-3], C13 H16 N3 NaO4 S, M r 333.34;
sodium salt monohydrate [50567-35-6],
C13 H16 N3 NaO4 S · H2 O, M r 351.36.
Synthesis [48, 211, 212] (see next page).
Clinical use: Nepafenac is a nonsteroidal an- Clinical use: Metamizol [213, 214] is the wa-
tiinflammatory drug, that was launched by Al- ter soluble sodium sulfonate of amidopyrine.
con in 2005 for the treatment of pain and inflam- After oral application it is rapidly hydrolyzed
mation associated with cataract surgery, because to 4-methylaminoantipyrine and metabolized to
it rapidly penetrates ocular tissues, and it is used various active metabolites [215]. Metamizol has
locally as 1 % ophthalmic suspensions [204]. strong analgesic, spasmolytic, and antipyretic,
Nepafenac is an amide analog and a pro- but no anti-inflammatory properties. The exact
drug of amfenac (2-amino-3-benzoylbenzene- mechanism of action is unknown but may in-
acetic acid), which is a COX-1 and COX-2 in- clude inhibition of prostaglandin synthesis. In-
hibitor [205]. Nepafenac is the first ophthalmic hibition of both COX isoenzymes by metami-
NSAID prodrug to receive FDA approval. It had zol has been demonstrated only when very high
originally been developed at Wyeth Consumer concentrations of this drug were used, thus ques-
Healthcare [204, 206, 207]. tioning the relevance of this activity.
Trade name: Nevanac. COX inhibition may be induced by the
metabolites, but this is not yet systematically in-
Amfenac [51579-82-9], 2-amino-3-ben- vestigated. Metamizol is used for the treatment
zoylbenzeneacetic acid, 2-amino-3-benzoyl- of medium to severe pain, often in combina-
phenylacetic acid, C15 H13 NO3 , M r 255.28, mp tion with opioids, for fever reduction and for
121-123 ◦ C (dec.); sodium salt monohydrate the treatment of colic pain. It is given by mouth
[61618-27-7], C15 H12 NNaO3 H2 O, M r 295.27, in doses of 500 mg up to 4 g daily, by intra-
mp 254-255.5 ◦ C. venous or by rectal application. Metamizol is rel-
Synthesis: The reaction of 1-aminoindolin- atively free of acute side effects but in rare cases
2-one with phenylacetone in presence of may cause severe and life-threatening allergic
28 Analgesics and Antipyretics

reactions like agranulocytosis, allergic skin re- Propyphenazone [479-92-5], 1,2-dihydro-


actions, and allergic shock. The compound is 1,5-dimethyl-4-(1-methylethyl)-2-phenyl-3H-
not used in the UK, USA, and in Scandinavian pyrazol-3-one, C14 H18 N2 O3 , M r 230.31, mp
countries. 103 ◦ C.
Synthesis [48, 216, 217]:

Clinical use: Propyphenazone [218, 219] is a


derivative of phenazone and has a similar anal-
gesic and antipyretic action. It is used as a mono-
compound as well as in multi-ingredient prepa-
rations.
Trade name: Demex (Germany).
Multi-ingredient preparation: Saridon (Ger-
many).

2.7. Acidic Enolic Compounds

2.7.1. Pyrazolidine-3,5-diones

Kebuzone [853-34-9], 4-(3-oxobutyl)-1,2-


Trade names: Analgin, Novalgin (Germany, diphenyl-pyrazolidine-3,5-dione, C19 H18 N2 O3 ,
Austria, Switzerland). M r 322.36, mp 115.5–116.5 ◦ C or 127.5–
Metamizol is also marketed in several generic 128.5 ◦ C, depending on crystal form.
formulations. Synthesis [158, 220, 221]:
Analgesics and Antipyretics 29

Clinical use: Mofebutazone [225] is a non-


steroidal anti-inflammatory drug used as sodium
salt for the treatment of mild to moderate
pain including inflammatory and degenerative
rheumatic disorders and musculoskeletal pain.
Mofebutazone can be given by oral, rectal
(900–1200 mg/d), or intramuscular administra-
tion (650 mg/d).
Trade name: Mofesal (Germany).

Oxyphenbutazone [129-20-4], oxyphenyl-


butazone, 4-butyl-1-(4-hydroxyphenyl)-2-
phenylpyrazolidine-3,5-dione, C19 H2 ON2 O3 ,
M r 324.37, mp 124–125 ◦ C; monohydrate
[7081-38-1], C19 H20 N2 O3 · H2 O, M r 342.40,
mp 96 ◦ C.
Synthesis [48, 226]:

Clinical use: Kebuzone [222] is a non-


steroidal anti-inflammatory drug used for the
treatment of acute and chronic pain and inflam-
mation states like musculoskeletal, joint, and
soft-tissue disorders. Kebuzone is administered
orally, rectally, or intramuscular (up to 1500
mg/d initial dose, 250–500 mg/d maintenance
dose).
Trade names: Ketazon (Austria), Chetopir
(Italy).

Mofebutazone [2210-63-1], 4-butyl-1- Clinical use: Oxyphenbutazone is a non-


phenylpyrazolidine-3,5-dione, C13 H16 N2 O2 , steroidal anti-inflammatory drug, which was
M r 232.28, mp 102–103 ◦ C; sodium salt used by oral, rectal, or topical administration
[41468-34-2], C13 H15 N2 NaO2 , M r 254.27. (400 - 600 mg/d) for the acute treatment of anky-
Synthesis [48, 223, 224]: losing spondylitis, chronic polyarthritis, and
gout. Because of a high incidence of severe side
effects including disturbances of the hematopoi-
etic system like agranulocytosis and aplastic
anemia [233] the compound is no longer used.
Oxyphenbutazone is a metabolite of phenyl-
butazone.

Phenylbutazone [50-33-9], 4-butyl-1,2-


diphenylpyrazolidine-3,5-dione, C19 H20 N2 O2 ,
30 Analgesics and Antipyretics

M r 308.37, mp 105 ◦ C; sodium salt [129-18-0], 335.34, mp 265–271 ◦ C (decomp.); sodium salt,
C19 H19 N2 NaO2 , M r 331.36; calcium salt (2 : 1) C14 H12 N3 NaO5 S, M r 343.29, mp 270–272 ◦ C.
[36298-23-4], C38 H36 CaN4 O4 , M r 656.83; pi- Synthesis [48, 231–233]:
perazine salt (1 : 1) [4985-25-5], C19 H20 N2 O2 ·
C4 H10 N2 , M r 395.51, mp 140–141 ◦ C.
Synthesis [48, 228]:

Clinical use: Phenylbutazone [229] is a


nonsteroidal anti-inflammatory drug used for
the acute treatment of ankylosing spondyli-
tis, chronic polyarthritis, and gout. Because of
severe side effects including disturbances of
the hematopoietic system like agranulocytosis
and aplastic anemia [230] the use is limited
to conditions in which other nonsteroidal anti-
inflammatory drugs do not show sufficient ef-
ficacy. Phenylbutazone is given as oral, rectal,
intramuscular or topical formulation (up to 600
mg/d initial dose, up to 400 mg/d maintenance
dose). The peak plasma concentration is reached
2 h after oral application. Phenylbutazone is
bound to 98 % to plasma protein. Oxyphenbuta- Clinical use: Isoxicam [234] is a nonsteroidal
zone is formed as an active metabolite of phenyl- anti-inflammatory drug withdrawn from the
butazone. market as a consequence of reports of fatal skin
Trade names: Butazolidin (France, Germany, reaction.
UK, USA), Ambene (Germany).
Lornoxicam [70374-39-9], 6-chloro-
4-hydroxy-2-methyl-1,1-dioxo-1,2-dihydro-
2.7.2. Arylsulfonamides (Oxicames) 1λ6 -thieno[2,3-e][1,2]thiazine-3-carboxyl-
ic acid pyridin-2-ylamide, 6-chloro-4-hy-
The oxicams are a group of nonsteroidal droxy-2-methyl-N-(2-pyridyl)-2H-thieno[2,3-
anti-inflammatory agents containing a 2H-1,2- e]-1,2-thiazine-3-carboxamide 1,1-dioxide,
benzothiazine-3-carboxamide 1,1-dioxide moi- C13 H10 ClN3 O4 S2 , M r 371.81, mp 225–230 ◦ C
ety as common structural entity (for Meloxicam (decomp.).
see Section 2.9.1). Synthesis: Sulfonation of 2,5-di-
chlorothiophene with ClSO3 H/SOCl2 gives
Isoxicam [34552-84-6], 4-hy- 2,5-dichlorothiophene-3-sulfonyl chloride,
droxy-2-methyl-1,1-dioxo-1,2-dihydro-1λ6 - which by reaction with methylamine in
benzo[e][1,2]thiazine-3-carboxylic acid CHCl3 yields the corresponding methy-
isoxazo-3-ylamide, 4-hydroxy-2-methyl-N-(5- lamide. The carboxylation with butyl lithium
methyl-3-isoxazolyl)-2H-1,2-benzothiazine-3- and CO2 in ether affords 5-chloro-3-(N-
carboxamide 1,1-dioxide, C14 H13 N3 O5 S, M r methylsulfamoyl)thiophene-2-carboxylic acid,
Analgesics and Antipyretics 31

which is esterified with PCl5 and metha- interleukin-6, both of which could contribute
nol to the methyl ester. Condensation with to its potent anti-inflammatory and analgesic
methyl iodoacetate by means of NaH in action [237]. It is used orally (8–24 mg/d) for
DMF gives methyl 5-chloro-3-[N-(methoxycar- the treatment of mild to moderate pain includ-
bonylmethyl)-N-methylsulfamoyl]thiophene- ing postoperative pain, rheumatoid arthritis, os-
2-carboxylate, which is cyclized with sodium teoarthritis, and ankylosing spondylitis [238].
methoxide in methanol yielding methyl 6- Lornoxicam shows a high binding to plasma
chloro-4-hydroxy-2-methyl-2H-thieno[2,3-e]- protein and in contrast to other oxicams a short
1,2-thiazine-3-carboxylate-1,1-dioxide. Finally plasma half-life of 3 to 5 h [236].
this compound is treated with 2-aminopyridine Trade names: Telos (Germany), Taigalor
in refluxing xylene [235]. (Italy), Xefo (Austria, Denmark, Italy, Switzer-
land, UK).

Piroxicam [36322-90-4], 4-hydroxy-


2-methyl-1,1-dioxo-1,2-dihydro-1λ6 -benzo-
[e][1,2]thiazine-3-carboxylic acid pyridin-2-
ylamide, 4-hydroxy-2-methyl-N-2-pyridinyl-
2H-1,2-benzothiazine-3-carboxamide 1,1-di-
oxide, C15 H13 N3 O4 S, M r 331.35, mp 198–
200 ◦ C.
Synthesis [239–243, 251]:

Clinical use: Piroxicam [244] is a non-


steroidal anti-inflammatory drug used for the
treatment of mild to moderate acute and chronic
pain and inflammation including musculoskele-
tal, soft-tissue, and joint disorders like ankylos-
ing spondylitis, chronic polyarthritis, and gout
[245]. Piroxicam shows a 600-fold selectivity
for COX-1 compared to COX-2 in cultured
animal cells [246]. Piroxicam is administered
orally, rectally, intramuscular, or topically (10–
30 mg/d, maximal initial dose 40 mg/d) as the
free base, as complex with beta-cyclodextrine,
Clinical use: Lornoxicam [236] is a non- and as cinnamate or pivalate. After oral admin-
steroidal anti-inflammatory drug with a strong istration, piroxicam reaches peak plasma con-
and balanced inhibition of COX-1 and COX-2 in centration after 3 to 5 h, shows a 99 % binding
cultured human cells [23]. In addition, lornoxi- to plasma protein and a long half-life of about
cam inhibits inducible nitric oxide synthase and 50 h.
32 Analgesics and Antipyretics

Trade names: Brexidol (EU, USA), Brexin 3-sulfothiophene-2-carboxylate, which by re-


(Franc, Italy), Feldéne (France), Felden (Ger- action with refluxing SOCl2 yields methyl-
many), Feldene (UK, USA). 3-chlorosulfonylthiophene-2-carboxylate. The
Piroxicam is also marketed in several generic following condensation with sarcosine ethyl
formulations. ester in hot CHCl3 gives 3-(N-ethoxycarbon-
ylmethyl-N-methylsulfamoyl)thiophene-2-car-
Tenoxicam [59804-37-4], 4-hydroxy-2- boxylate, which is cyclized by treatment with
methyl-1,1-dioxo-1,2-dihydro-1λ6 -thieno[2,3- sodium methoxide in refluxing methanol afford-
e][1,2]thiazine-3-carboxylic acid pyridin-2- ing 3-ethoxycarbonyl-4-hydroxy-2-methyl-2H-
ylamide, 4-hydroxy-2-methyl-N-2-pyridinyl- thieno-[2,3-e]-1,2-thiazine 1,1-dioxide. Finally
2H-thieno[2,3-e]-thiazine-3-carboxamide 1,1- this compound is condensed with 2-aminopyri-
dioxide, C13 H11 N3 O4 S2 , M r 337.38, mp 209– dine in refluxing toluene [48, 243, 247].
213 ◦ C (decomp.). Clinical use: Tenoxicam [248] is a non-
steroidal anti-inflammatory drug used for the
treatment of mild to moderate pain states in
musculoskeletal, soft-tissue, and joint disorders
like rheumatoid arthritis, osteoarthritis and gout.
Tenoxicam shows a balanced inhibition of both
COX-1 and COX-2 in cultured human cells [23].
Tenoxicam is administered orally, rectally, or in-
tramuscular (20 mg/d, maximal dose 40 mg/d).
Peak plasma concentration appears within 1 to 6
h after administration depending on food intake.
Tenoxicam is bound to plasma protein to 98.5 %
and shows a long plasma elimination half-life of
70 to 90 h.
Trade names: Mobiflex (UK), Tilcotil
(France, Italy, Switzerland).

2.8. Other Structures


Ketorolac [74103-06-3], 5-benzo-
yl-2,3-dihydro-1H-pyrrolizine-1-carboxylic
acid, C15 H13 NO3 , M r 255.27, mp 160–
161 ◦ C; tromethamine salt (1 : 1) [74103-07-
4], C15 H13 NO3 · C4 H11 NO3 , M r 376.41, (+)-
form mp 174 ◦ C, [α]D + 173◦ (c = 1, CH3 OH),
(−)-form mp 169–170 ◦ C, [α]D − 176◦ (c =
1, CH3 OH); monosodium salt [110618-38-7],
C15 H12 NNaO3 , M r 277.26.
Synthesis: Benzoylation of 2-methyl-
thiopyrrole with N,N-dimethylbenzamide in the
presence of POCl3 in refluxing CH2 Cl2 gives
Synthesis: The reaction of methyl 3-hy- 5-benzoyl-2-methylthiopyrrole, which is con-
droxythiophene-2-carboxylate with PCl5 in re- densed with spiro[2,5]-5,7-dioxa-6,6-dimethyl-
fluxing CCl4 gives 3-chlorothiophene-2-car- octane-4,8-dione by means of NaH in DMF.
boxylic acid, which by treatment with NaHSO3 Oxidation of this product with m-chloroperben-
and Cu in aqueous alkaline solution at 143 ◦ C zoic acid in CH2 Cl2 affords the sulfone, which
in a pressure vessel is converted into 3- is submitted to methanolysis with methanol and
sulfothiophene-2-carboxylic acid. Its esterifi- HCl giving 1-(3,3-dimethoxycarbonylpropyl)-
cation with refluxing methanol affords methyl- 2-methanesulfonyl-5-benzoylpyrrole. Cycliza-
Analgesics and Antipyretics 33

tion with NaH in DMF yields dimethyl-5-benzo- the tromethamine salt. Due to a number of se-
yl-1,2-dihydro-3H-pyrrolo[1,2-α]pyrrole-1,1- vere side effects including gastrointestinal dis-
dicarboxylate, which is finally hydrolyzed and turbances, impairment of liver functions, renal
decarboxylated with KOH in refluxing methanol failures, skin irritations, and other hypersensi-
[48, 249–253]. tivity reaction it has been withdrawn in many
countries.
Trade names: Acular (UK, USA), Droal
(Spain), Tora-Dol (France, Italy), Toradol (UK,
USA).

Oxaprozine [21256-18-8], 3-(4,5-diphen-


yl-oxazol-2-yl)propionic acid, C18 H15 NO3 , M r
293.32, mp 160.5–161.5 ◦ C.
Synthesis [255–257]:

Clinical use: Oxaprozine [257] is a non-


steroidal anti-inflammatory drug used for the
treatment of mild to moderate pain includ-
ing rheumatoid arthritis and osteoarthritis.
Oxaprozine shows slow elimination kinetics
with an elimination half-life of about 24 h. It
is given orally (600–1200 mg/d, maximum dose
1800 mg/d).
Trade names: Deflam (South Africa), Daypro
(USA).

2.9. COX-2 Inhibitors

2.9.1. Nonselective COX-2 Inhibitors


Clinical use: Ketorolac [254] is a non-
steroidal anti-inflammatory drug mainly used Etodolac [41340-25-4], (1,8-diethyl-
for the treatment of moderate to severe post- 1,3,4,9-tetrahydropyrano[3,4-b]indol-1-yl)-
operative pain. Ketorolac shows a balanced in- acetic acid, C17 H21 NO3 , M r 287.35, mp 145–
hibition of COX-1 and COX-2 in cultured hu- 148 ◦ C.
man cells [23]. Ketorolac is mostly used as Synthesis [258–261]:
34 Analgesics and Antipyretics

Synthesis: Reaction of benzothiazolo-3(2H)-


one-1,1-dioxide with methyl chloroacetate gives
the methyl 2(3H)-acetate derivative, which is
isomerized with sodium methoxide in toluene-
tert-butanol yielding methyl 4-hydroxy-2H-1,2-
benzothiazine-3-carboxylate-1,1-dioxide. Sub-
sequent methylation with methyl iodide in
methanol yields the 2-methyl compound. Fi-
nally this compound is treated with 2-amino-5-
methylthiazole in xylene [48, 265, 266].

Clinical use: Etodolac [262] shows a ten-fold


selectivity for COX-2 compared to COX-1 in
human whole blood and belongs to the first gen-
eration of COX-2 inhibitors [22]. Etodolac is a
racemate with an active (S)-enantiomer and an
inactive (R)-enantiomer. It is used for the treat-
ment of mild to moderate acute and chronic pain Clinical use: Meloxicam [267] shows a ten-
including rheumatoid arthritis and osteoarthri- fold selectivity for COX-2 compared to COX-
tis. Clinical data indicate fewer gastrointesti- 1 in human whole blood and belongs to the
nal side-effects in comparison to naproxen [51, first generation of COX-2 selective drugs [22].
264]. Etodolac is used in oral slow and immedi- Meloxicam is used for the acute and chronic
ate release preparations and applied in doses of treatment of mild to moderate pain including
600–1200 mg/d. Peak plasma concentrations are arthritis and ankylosing spondylitis. Meloxi-
reached within 2 h. Etodolac shows a 99 % bind- cam shows central antinociceptive effects in rats
ing to plasma protein and an elimination half-life which seems to be independent from the COX
of about 7 h. inhibiting properties [268].
Trade name: Lodine (USA, UK, France). Meloxicam is given by oral administration
(7.5–15 mg/d). It shows an elimination half-life
Meloxicam [71125-38-7], 4-hy- of 20 h.
droxy-2-methyl-1,1-dioxo-1,2-dihydro-1λ6 - Trade names: Mobec (Germany), Mobic
benzo[e][1,2]thiazine-3-carboxylic acid (5- (Italy, UK, USA), Movalis (Spain).
methyl thiazol-2-yl)amide, 4-hydroxy-
2-methyl-N-(5-methyl-2-thiazoyl)-2H-1,2- Nimesulide [51803-78-2], N-(4-
benzothiazine-3-carboxamide 1,1-dioxide, nitro-2-phenoxyphenyl)methanesulfonamide,
C14 H13 N3 O4 S2 , M r 351.40, mp 264 ◦ C (de- C13 H12 N2 O5 S, M r 308.05, mp 143–144.5 ◦ C.
comp.). Synthesis [48, 269]:
Analgesics and Antipyretics 35

ing methanol using sodium methoxide as


catalyst gives 4,4,4-trifluoro-1-(4-methylphe-
nyl)butane-1,3-dione, which is cyclized with 4-
hydrazinophenylsulfonamide in refluxing etha-
nol [488, 493].

Clinical use: Nimesulide [270] is a first-


generation COX-2 inhibitor with a 5- to 100-
fold selectivity for COX-2 compared to COX-1,
depending on the assay system [22, 23]. It is Clinical use: Celecoxib [166] is a second-
used for the short-term treatment of inflamma- generation selective COX-2 inhibitor and was
tory conditions, fever, and pain, including mus- the first drug of this group to reach the market.
culoskeletal and joint disorders. In addition to The selectivity for COX-2 compared to COX-1
COX inhibition nimesulide reduces the produc- is about 375-fold in human recombinant enzyme
tion of the pro-inflammatory cytokine TNF-α preparations and about eight-fold in a whole
under inflammatory conditions [271]. blood assay. Celecoxib has been approved in
Nimesulide is available in oral or rectal for- 1999 in the USA and is now used for the treat-
mulations (up to 400 mg/d). ment of rheumatoid arthritis and osteoarthritis,
The use of nimesulide has been reported to ankylosing spondylitis, acute pain and primary
be associated in some cases with hepatic failure dysmenorrhoea all over the world.
[272, 273] and the compound was withdrawn Celecoxib is further approved for the use in
from the market in several countries. familial adenomatous polyposis in the US and
Trade names: Aulin (Italy, Switzerland), leads to a reduction in the number of colorectal
Nexen (France), Guaxan (Spain, withdrawn). polyps in these patients [276].
The use of celecoxib is associated with a
reduced incidence of gastroduodenal ulcers in
2.9.2. Selective COX-2 Inhibitors (Coxibs) comparison to naproxen [277], ibuprofen, and
diclofenac [278] in arthritic patients. Because of
Celecoxib [169590-42-5], 4-(5-p-tolyl- a greater COX-1 inhibiting component as com-
3-trifluoromethylpyrazol-1-yl)-benzenesulfon- pared to the newer highly COX-2 selective com-
amide, C17 H14 F3 N3 O2 S, M r 381.37, mp 157– pounds such as valdecoxib, rofecoxib, and pari-
159 ◦ C (pale yellow solid). coxib, cardiovascular side effects of celecoxib
Synthesis: Condensation of 4-methylaceto- are less prominent and the compound avoided
phenone with ethyl trifluoroacetate in reflux- withdrawal from the market [279]. Nevertheless
36 Analgesics and Antipyretics

celecoxib should be used with appropriate pre- 5-chloro-3-[4-(methylsulfonyl)phenyl]-


caution. pyridin-2-amine, which is converted
Plasma peak concentrations are achieved into 2,5-dichloro-3-[4-(methylsulfo-
within 2 h and the elimination half-life is about nyl)phenyl]pyridine by treatment first with
12 h. Celecoxib is given orally (200–400 mg/d). NaNO2 and HCl and then chlorination
Trade names: Celebrex (EU, USA), Onsenal with POCl3 [280, 281].
(EU). iii) 2,5-dichloro-3-[4-(methylsulfonyl)phenyl]-
pyridine is condensed with either trimeth-
Etoricoxib [202409-33-4], [202409-40- yl(6-methyl-3-pyridyl)tin or the boronate
3] (mono HCl), 5-chloro-3-[4-(methyl-sul- ester lithium salt by means of Pd(PPh3 )4 to
fonyl)phenyl]-2-(6-methylpyridin-3-yl)pyri- afford etoricoxib .The metalated pyridine
dine, 5-chloro-6 -methyl-3-(4-[methylsulfo- 2-methyl-5-(trimethylstannyl)pyridine is
nyl)phenyl]-2,3 -bipyridine, C18 H15 ClN2 O2 S, obtained by esterification of 3-hydroxy-6-
M r 358.848, crystals, mp 271.5-138.1 ◦ C methylpyridine with trifluoromethanesul-
(136.7 ◦ C DSC onset). fonic (triflic) anhydride to give the corre-
Synthesis [280–284]: sponding triflate 6-methylpyridin-3-yl tri-
fluoromethanesulfonate, which is treated
a) Synthesis via dichloropyridine: with hexamethylditin to afford the tar-
i) The bromination of 5-chloro-2-hy- get tin intermediate 2-methyl-5-(trimeth-
droxypyridine with Br2 in acetic acid ylstannyl)pyridine. The boronate lithium
gives 3-bromo-5-chloro-2-hydroxypyri- salt is prepared by treatment of 5-bromo-
dine, which is treated with benzyl bro- 2-methylpyridine with n-butyl lithium fol-
mide and Ag2 CO3 in hot benzene to lowed by addition of triisopropyl borate
yield the benzyl ether 2-(benzyloxy)-3- [281] (see next page).
bromo-5-chloropyridine. Condensation of b) In addition, etoricoxib is obtained by sev-
2-(benzyloxy)-3-bromo-5-chloropyridine eral related ways via the ketosulfone key
with 4-(methylsulfanyl)phenylboronic intermediate 1-(6-methylpyridin-3-yl)-2-
acid with the aid of Pd(PPh3 )4 and [4-(methylsulfonyl)phenyl]ethanone, which
Na2 CO3 in refluxing ethanol/benzene can also be synthesized by several different
affords 2-(benzyloxy)-5-chloro-3-[4- routes:
(methylsulfanyl)phenyl]pyridine, which i) Reaction of ketosulfone 1-(6-
is oxidized with OsO4 and sodium sul- methylpyridin-3-yl)-2-(4-[methylsulfo-
fite to furnish sulfone 2-(benzyloxy)- nyl)phenyl]ethanone with 2-chloro-1,3-
5-chloro-3-[4-(methylsulfonyl)phenyl]- bis(dimethylamino)trimethinium hexa-
pyridine. Treatment of 2-(benzyloxy)-5- fluorophosphate salt in the presence of
chloro-3-[4-(methylsulfonyl)phenyl]pyri- an equimolar amount of potassium tert-
dine with trifluoroacetic acid (TFA) pro- butoxide, followed by treatment with
vides the 2-hydroxypyridine derivative, acetic/trifluoroacetic acid and then heating
which is reacted with POCl3 to yield 2,5- at reflux with an excess of ammonium hy-
dichloro-3-[4-(methylsulfonyl)phenyl]- droxide. The 2-chloro-1,3-bis(dimethyl-
pyridine. amino)trimethinium hexafluorophosphate
ii) Bromination of 2-amino-5-chloropyr- salt is obtained by reaction of chloroacetic
idine with Br2 in acetic acid pro- acid with hot dimethylformamide and
vides 2-amino-3-bromo-5-chloropyr- POCl3 , and then the reaction mixture
idine, which is condensed with 4- is treated with 5 N NaOH and hexa-
(methylsulfanyl)phenylboronic acid in fluorophosphoric acid in water.
the presence of Pd(PPh3 )4 and Na2 CO3 ii) Cyclization of ketosulfone 1-(6-
in refluxing ethanol/benzene to give 5- methylpyridin-3-yl)-2-(4-[methyl-
chloro-3-[4-(methylthio)phenyl]pyridin- sulfonyl)phenyl]ethanone with 2-
2-amine. Oxidation of 5-chloro-3-[4- chloromalondialdehyde (A: X=OH),
(methylthio)phenyl]pyridin-2-amine with or by cyclization of ketosulfone 1-(6-
OsO4 as before yields the sulfone methylpyridin-3-yl)-2-[4-(methylsulfo-
Analgesics and Antipyretics 37

nyl)phenyl]ethanone with aminoacrolein treatment of chloromalondialdehyde with


(B: X=NH2 ) in the absence of am- oxalyl chloride and dimethyl formamide in
monium acetate. The cyclization of toluene – followed by reaction with am-
the lithium enolate of ketosulfone monium acetate or anhydrous ammonia
1-(6-methylpyridin-3-yl)-2-(4-(methyl- yield also the desired etoricoxib.
sulfonyl)phenyl)ethanone with 2,3-di- iii) Etoricoxib can also be obtained by cycliza-
chloroacrolein (C: X=Cl) – obtained by tion of the aniline derivative 2-chloro-3-
38 Analgesics and Antipyretics

phenylaminopropenal and ammonium ac- Lumiracoxib [220991-20-8], 2-[2-(2-


etate in hot propionic acid [280–284]. chloro-6-fluorophenylamino)-5-methylphe-
Clinical use: Etoricoxib [280, 285] is a third- nyl]acetic acid, C15 H13 ClFNO2 , M r 293.72,
generation COX-2 inhibitor with a 100-fold se- mp 158-159 ◦ C.
lectivity for COX-2 in a whole blood assay. The Synthesis [61, 294, 295]: The partial reduc-
selectivity ratio is higher than with other coxibs tion of 4-methylanisole with sodium in liquid
such as rofecoxib, valdecoxib, or celecoxib. ammonia/THF/ethanol gives the enol ether 3-
The compound was shown to be active in methoxy-6-methylcyclohexa-1,4-diene, which
rheumatoid arthritis, osteoarthritis, ankylosing is condensed with 2-chloro-6-fluoroaniline in
spondylitis, postoperative pain, and in chronic presence of TiCl4 in chlorobenzene/THF to
low back pain. Further positive results were seen yield (E)-2-chloro-6-fluoro-N-(4-methylcyclo-
in patients with acute gout and against primary hex-2-enylidene)benzenamine, which, without
dysmenorrhoea [286]. isolation, is aromatized with I2 in AcOH/THF
Etoricoxib is used in oral doses of 60–120 to provide N-(2-chloro-6-fluorophenyl)-N-(4-
mg/d. The compound is rapidly absorbed and methylphenyl)amine. The acylation of this
peak plasma concentrations are achieved within intermediate with 2-chloroacetyl chloride at
1.0 to 1.5 h after administration in healthy vol- 90 ◦ C affords the 2-chloro-N-(2-chloro-6-
unteers [287]. The elimination half-life is about fluorophenyl)-N-p-tolylacetamide, which is
15 h. Etoricoxib is metabolized to about 60 % cyclized by means of AlCl3 by heating at
by members of the cytochrome P450 3A family 160 ◦ C to afford 1-(2-chloro-6-fluorophenyl)-
[288]. In vitro studies give no evidence for active 5-methylindolin-2-one. Finally, this compound
metabolites with respect to COX-1 and COX-2 is hydrolyzed with NaOH in refluxing etha-
inhibition [289]. nol/water and acidified with 1 N HCl to give
Etoricoxib is generally well tolerated and in- lumiracoxib. Alternatively, the intermediate
duces less gastrointestinal side effects than con- N-(2-chloro-6-fluorophenyl)-N-(4-methylphe-
ventional COX-1 inhibitors or non-COX selec- nyl)amine can also be obtained by condensa-
tive NSAIDs [290]. Renal tolerability is simi- tion of 2-chloro-N-(4-methylphenyl)acetamide
lar to conventional COX-1 inhibitors [291]. Ac- with 2-chloro-6-fluorophenol using K2 CO3 in
cording to its COX-2 selectivity, etoricoxib has isopropanol to yield 2-(2-chloro-6-fluorophen-
an increased risk of cardiovascular side effects oxy)-N-(4-methylphenyl)acetamide, which is
[292] and should only be used with appropriate treated with MeONa in methanol to obtain the
precautions. target secondary amine Lumiracoxib [61, 294].
Trade name: Arcoxia (EU, USA).
Analgesics and Antipyretics 39

acetic acid. Reaction of 2-(2-iodo-5-methylphe-


nyl)acetic acid with SOCl2 in refluxing dichloro-
methane gives the corresponding acyl chlo-
ride, which is treated with dimethylamine in
diethyl ether/THF to yield 2-(2-iodo-5-meth-
ylphenyl)-N,N-dimethylacetamide. Condensa-
tion of this intermediate with 2-chloro-
6-fluoroaniline by means of Cu powder,
Cu2 I2 and K2 CO3 in refluxing xylene af-
fords 2-[2-(2-chloro-6-fluorophenylamino)-5-
methylphenyl]-N,N-dimethylacetamide, which
is finally hydrolyzed with NaOH in refluxing bu-
tanol/water to give lumiracoxib [61, 294, 295].

An alternative synthesis of lumiracoxib


involves the coupling of N,N-dimethyl-5-
methyl-2-iodophenylacetamide with 2-chloro-
6-fluoroaniline in the presence of copper pow-
der, copper(I) iodide and anhydrous potassium Clinical use: Lumiracoxib [296] is a novel
carbonate, and refluxing in xylene for 48 h. COX inhibitor with the highest COX-2 selectiv-
This synthetic route includes the following ity of all coxibs. The compound is structurally
steps: The reduction of 2-iodo-5-methylbenzo- related to the phenylacetic acid COX inhibitors,
ic acid with BH3 /THF in THF gives 2-iodo- has weakly acidic properties and is devoid of
5-methylbenzyl alcohol, which is treated with the sulfonamide or methylsulfone group of the
refluxing 48 % HBr to yield the benzyl bro- other coxibs. The compound showed clinical ef-
mide. Reaction of the benzyl bromide with ficacy against osteoarthritis [297] and rheuma-
NaCN in ethanol/water affords the phenylace- toid arthritis [298] and inhibited acute pain re-
tonitrile, which is hydrolyzed with NaOH in lated to primary dysmenorrhoea, dental or ortho-
refluxing EtOH/water to provide the phenyl- pedic surgery, or tension-type headache [299].
40 Analgesics and Antipyretics

The compound is well tolerated and gas- (valdecoxib), with propionic anhydride in a so-
trointestinal side effects were significantly lower lution of triethanolamine (TEA) and 4-dimeth-
than after treatment with COX-1 inhibitors like ylaminophenol (DMAP) in tetrahydrofuran
ibuprofen or naproxen [300, 301] The meta- gives N-[4-(5-methyl-3-phenylisoxazol-4-
analysis of all clinical trials in osteoarthritis and yl)phenylsulfonyl]propionamide, which is
rheumatoid arthritis gave no indication of an in- treated with NaOH in ethanol to give the sodium
creased risk of cardiovascular side effects [302]. salt of parecoxib [79, 306, 307].
Liver function tests showed a moderate and re- Clinical use: Parecoxib [77, 78, 80] is a third-
versible increase in liver enzymes [303]. generation COX-2 inhibitor. Parecoxib is a pro-
Lumiracoxib has a high oral bioavailability drug of valdecoxib with aqueous solubility suffi-
of about 74 %, is rapidly absorbed, reaches its cient for the use in parenteral formulations. The
peak plasma concentrations within 2 h and is compound induced potent pain inhibition in den-
strongly bound to plasma protein. The com- tal, gynecological and orthopedic surgery and
pound is strongly metabolized, is devoid of ac- produced less gastrointestinal side effects than
tive metabolites and has a plasma elimination conventional COX-1 inhibitors [81].
half-life of about 4 h [304]. Despite the short Parecoxib is rapidly hydrolyzed in the liver
half-life, a once-daily administration in doses of to its active metabolite valdecoxib. Plasma
100–400 mg is recommended. A dose of 100 mg peak concentrations for valdecoxib are achieved
is used for the symptomatic relief of osteoarthri- within 1.1 to 3.5 and 0.27 to 2 h after i.m.
tis and 400 mg for short-term relief of moderate and i.v. administration, respectively. The elim-
to severe pain, associated with dental or ortho- ination half-life for parecoxib is 15 to 35 min
pedic surgery. As with other COX-2 inhibitors, and 5 min for i.m. and i.v. administration, re-
the compound should be used with appropriate spectively. Metabolism of parecoxib follows the
precaution. metabolism of the active metabolite valdecoxib
Trade name: Prexige (EU, USA). which is a substrate for CYP3A4 and CYP2C9.
Parecoxib was used in doses of 20 and 40 mg
Parecoxib [198470-84-7], (free acid) of its sodium salt in the short-term treatment of
[198470-85-8], N-(4-(5-methyl-3-phenyl- postoperative pain.
isoxazol-4-yl)phen-ylsulfonyl)propionamide Parecoxib together with valdecoxib was with-
sodium salt C19 H17 N2 O4 SNa, M r 392.409, drawn from the market because of an increased
crystals, mp 271.5–272.7 ◦ C. risk of cardiovascular side effects [312, 313].
Trade names: Dynastat, Rayzon, Xapit (com-
pound withdrawn from EU and US markets).

Rofecoxib [162011-90-7], 4-[4-(methyl-


sulfonyl)phenyl]-3-phenylfuran-2(5H)-one,
C17 H14 O4 S, M r 314.36.
Synthesis: Condensation of phenylacetic
acid with ethyl bromoacetate in the pres-
ence of triethylamine in THF yields ethyl
2-(phenylacetoxy)acetate, which is cyclized
to the hydroxyfuranone using potassium
tert-butoxide in tert-butanol. The reaction
with trifluorosulfonyl anhydride and diiso-
propylethylamine in CH2 Cl2 affords the
corresponding trifluorosulfonate, which by
reaction with LiBr in hot acetone yields
the bromofuranone. The coupling with 4-
(methylsulfanyl)phenylboronic acid with
Na2 CO3 and Pd[(C6 H5 )3 P]4 as catalysts in hot
Synthesis: The acylation of 4-(5-methyl- toluene gives 4-[4-(methylsulfanyl)phenyl]-3-
3-phenylisoxazol-4-yl)benzenesulfonamide phenylfuran-2(5H)-one, which is finally oxi-
Analgesics and Antipyretics 41

dized with 2KHSO5 · KHSO4 · K2 SO4 (oxone) tion of dibutylurea (DBU) effected the cycliza-
[314]. tion to provide rofecoxib as the final product.

Clinical use: Rofecoxib [156, 317, 318] is a


second-generation selective COX-2 inhibitor. It
was the second COX-2 selective drug to reach
the market in 1999. The selectivity for COX-2
compared to COX-1 is more than 800-fold in
cellular assays and more than 10-fold in whole
blood assays [319].
Rofecoxib is used for treatment of rheuma-
toid arthritis, osteoarthritis (12.5–25 mg/d) and
pain of different origin such as postoperative
pain, dental pain, and primary dysmenorrhoea
(50 mg/d).
Rofecoxib reaches peak plasma concentra-
tions between 2 to 9 h after oral administra-
Rofecoxib can also synthesized by several tion. It is bound about 87 % to plasma protein
different routes [156]. In 2001 a highly ef- and has an elimination half-life of about 17 h.
ficient synthesis of rofecoxib was described Therefore a once daily dosing is appropriate.
[316]. As illustrated in the following scheme, The main metabolites formed in the liver are
the acetophenone (i) is prepared according the cis-dihydro and trans-dihydro derivatives of
to the literature by Friedel–Crafts acylation rofecoxib. Rofecoxib is excreted in the form
with thioanisole. Oxidation with magnesium of its metabolites mainly in the urine (72 %)
monoperoxyphthalate hexahydrate (MMPP) af- with some unchanged drug excreted in the fe-
fords the sulfone (ii), which is reacted with ces (14 %).
bromine in chloroform in the presence of a trace Rofecoxib shows significantly less gastroin-
amount of AlCl3 to give (iii). Bromoketone (iii) testinal toxicity than ibuprofen in studies with
is then coupled and cyclized in a second step, in osteoarthritis patients [320] and compared to
a one-pot procedure with phenylacetic acid. The naproxen in patients with rheumatoid arthri-
mixture of bromoacetophenone (iii) and phenyl- tis [321]. Rofecoxib does not prolong bleeding
acetic acid in acetonitrile was then treated with time.
triethylamine at room temperature to provide the Rofecoxib was withdrawn from the market at
ester intermediate, subsequent cooling and addi- the end of 2004 after it had been shown that use
for a period of more than 18 month increased
42 Analgesics and Antipyretics

the risk of heart attack and stroke [31]. It is be- Valdecoxib is converted in rodents and dogs,
ing discussed with the health authorities that a and in a low abundance in humans, by hy-
relaunch should be possible under appropriate droxylation of the methyl group to an ac-
safety regulations [322]. tive metabolite (4-(5-hydroxymethyl-3-phenyl-
Trade name: Vioxx (EU, USA; currently isoxazol-4-yl)benzenesulfonamide).
withdrawn from the market). Valdecoxib induced severe allergic skin re-
actions [330, 331] which, together with an in-
Valdecoxib [181695-72-7], 4-(5-methyl- creased risk of cardiovascular side effects [332]
3-phenylisoxazol-4-yl)benzenesulfonamide, was the reason why the compound, together with
C16 H14 N2 O3 S, M r 314.366; mp 172-173 ◦ C. its prodrug derivative parecoxib, was withdrawn
Synthesis: Deoxybenzoin is converted to from the market in 2004.
the corresponding oxime by treatment with Trade name: Bextra (EU, USA; currently
hydroxylamine under basic conditions with withdrawn from the market).
sodium acetate in aqueous ethanol or in toluene
in presence of potassium hydroxide in absolute
ethanol. The treatment of the oxime under nitro- 3. Centrally Acting Analgesics
gen with two equivalents of butyllithium in tetra-
hydrofuran is followed by cyclization in ethyl 3.1. Opioids
acetate or acetic anhydride to the isoxazoline
derivative. Finally, treatment of the isoxazoline The term “centrally acting analgesics” is used
with cold chlorosulfuric acid followed by reac- for compounds which inhibit the pain reaction
tion of the intermediate with aqueous ammonia predominantly within the central nervous sys-
affords the desired product [79–81, 156, 316, tem. Most of them induce a very powerful pain
323]. inhibition and are thus named synonymously
“strong analgesics”. The most important repre-
sentatives of this group are the opioids [333]. In
respect to structural features [334] opioids can
be separated into three groups:
• The first group contains the natural products
morphine, codeine, and thebaine, which are
isolated from opium. The group contains in
addition various semisynthetic derivatives of
the three compounds, which are prepared by
chemical modifications of these natural prod-
ucts.
• The second group encompasses fully syn-
thetic compounds which often have a to-
Clinical use: Valdecoxib [79, 324–326] is tal different chemical structure than the
a third-generation COX-2 inhibitor. It shows semisynthetic analogues, but interact with
about 30-fold selectivity for COX-2 in a whole- the same opioid receptors and show the same
blood assay. The compound has a very low sol- spectrum of analgesia and side effects as the
ubility in water and can only be administered natural compounds. For both groups together
orally. For parenteral administration, a water- the older name “opiates“ is still in use.
soluble prodrug derivative, parecoxib, was de- • The third group consists of naturally occur-
veloped in parallel. Valdecoxib showed strong ring and synthetic peptides with opioid-like
analgesic and anti-inflammatory properties and properties. The opioid peptides were disco-
was developed for the treatment of osteoarthritis, vered during the search for endogenous lig-
rheumatoid arthritis [327] and acute and chronic ands of the opioid receptors and share the
pain of various origin [328] including migraine same action and side effect profile with the
headache [329]. nonpeptidic compounds, but they are not in
clinical use.
Analgesics and Antipyretics 43

Opioids interact with specific receptors (re- receptor (ligand= ketazocine) and the σ-receptor
ceptor ligands) and they can act as pure agonists, (ligand = SKF 100 81). This was later confirmed
as partial agonists (agonists with a reduced in- by binding experiments with radioactive-labeled
trinsic activity) or as antagonist (binding without ligands and by the different binding and action
intrinsic activity). Only few compounds of the profiles of the endogenous opioid peptides, the
opioid family are selective ligands for a single enkephalins and endorphines [342]. Up to now,
type of opioid receptor (Table 3). Most of them three types of receptors, the µ-, κ-, and δ-opioid
bind with similar or different affinities to more receptor have been confirmed [343, 344], the σ-
than one receptor type and the pharmacological receptor is no longer considered to be an opioid
effect is the result of the combined effects on all receptor.
receptors involved. Opioids may act as a full or More recently, a fourth opioid receptor type,
partial agonist on one receptor type and as an named opioid receptor like (ORL1) receptor has
antagonist on the other. These compounds are been identified [345]. The receptor was detected
named mixed agonist – antagonists and they are as a c-DNA, which coded for a protein with
an important subgroup of opioid analgesics (see opioid receptor-like properties. Within a short
Section 3.1.2.1). Opioid analgesics are the most time, the endogenous ligand, a peptide named
important drugs for the treatment of moderate, nociceptin, was isolated, which, depending on
severe, and very severe pain. the place and route of application, induced pro-
Humans and most animal species are nociceptive or anti-nociceptive actions. The full
equipped with opioid receptors and endogenous spectrum of biological activity of nociceptin and
ligands. This endogenous opioid system [336] is the physiological role of the ORL-1 opioid re-
widely distributed within the body, it is phyllo- ceptor in pain processing is still under investi-
genetically very old and is expressed in all ver- gation [367].
tebrates. A high density of opioid receptors is
found in the brain [337] and in the spinal cord, Subtypes of the Different Opioid Recep-
where it is involved in pain inhibition and ad- tors. Corresponding to other receptor systems,
ditionally in many other central regulatory pro- binding studies and functional investigations in-
cesses. In addition to the CNS localization, opi- dicate that subtypes of opioid receptors exist
oid receptors are expressed in many peripheral [346, 347]. Within the µ-receptor two subtypes,
organs [338]. Of great importance are the opioid the µ-1 and µ-2 receptor have been described.
receptors of the gastrointestinal system, which It was postulated by some investigators [347]
regulate stomach emptying, gut motility, and that analgesia and opioid side effects are differ-
intestinal fluid secretion. Opioid receptors are ently distributed between µ-receptor subtypes,
found in cells of the immune system and periph- which should allow separation of analgesia from
eral opioids seem to be involved in the regulation the unwanted opioid side effects. According to
of inflammatory and immunological processes Pasternak and Wood [347], analgesia should be
[339]. In addition to the outstanding role in the mediated by the µ-1 receptor site, whereas res-
central pain inhibition, action at peripheral opi- piratory depression and addiction is mediated
oid receptors, which are expressed in high quan- via the µ-2 receptor subtype. But in contrast to
tities during inflammation and immune stimula- binding experiments, the functional separation
tion, may add to central pain inhibition [340]. of the µ-1 and µ-2 subtype is more equivocal
and a clear separation of analgesia from res-
Opioid Receptor Types. Differences in piratory depression and addiction potential had
analgesic activity and in the side effect pro- never been found within the µ-opioids. There-
file of synthetic opioids reinforced speculations fore, a differentiation of µ-opioid analgesics ac-
that more than one type of opioid receptor ex- cording to subtype specificity is no longer main-
ists and is involved in the analgesic activity of tained.
these compounds. Martin and coworkers inves- The subtypes of µ-opioid receptors could
tigated in 1960 these differences in a specially not be confirmed in cloning experiments [348].
developed test model, the chronic spinal dog Therefore, possible heterogeneity of opioid re-
[341] and postulated three types of opioid recep- ceptor subtypes must result from a later modifi-
tors, the µ-receptor (ligand = morphine), the κ- cation which is independent from the gene level.
44 Analgesics and Antipyretics

Possible variations could include splice variants, a novel opioid-like cDNA was isolated [351],
receptor association, posttranslational modifica- which coded for an unknown opioid receptor.
tions (e.g., glycosidation) or coupling with dif- The new receptor had many similarities with the
ferent transduction mechanisms. classical opioid receptors [352] and was added
as the forth member to the opioid receptor family
Endogenous Opioid Peptides. For each under the name ORL-1. All four opioid recep-
class of opioid receptors endogenous ligands tors have been identified in humans. All together,
of peptidic structure exist, which show a dis- they show a high degree of structural identity,
tribution within the body corresponding to that which corresponds to their widely overlapping
of the receptor. Three distinct families of lig- biological functions [348, 353].
ands with various members have been identi- All opioid receptors belong to the group
fied and named endorphins, enkephalins, and of pertussis toxin sensitive G-protein-coupled
dynorphins (Table 2). The opioid peptides are receptors of the rhodopsin family with seven
split off in the organism from specific precursor transmembrane spanning hydrophobic domains.
polypeptides. Enkephalins are derived from pro- The N-terminal part is oriented to the outer side
enkephalin and interact with µ- and δ-opioid re- of the cell membrane and is involved in the selec-
ceptors. The precursor molecule of endorphin is tion and binding of the receptor specific ligands.
pro-opiomelanocortin (POMC), which addition- Studies with chimeric or point mutated recep-
ally releases peptidic hormones such as α-MSH, tors indicate that predominantly the second and
ACTH and β-LPH. Endorphins are specific lig- third extracellular loop determines receptor se-
ands of the µ-opioid receptor type. Dynorphins lectivity. The N-terminal sequence contains sev-
interact with the κ-opioid receptor and are de- eral free amino groups which can be conjugated
rived from pro-dynorphin. ORL binding sites with sugar residues. The carboxy-terminal is di-
have recently been identified together with its rected into the inner part of the cell and is in-
endogenous peptidic ligand named nociceptin volved in the signal transduction cascade. The
(Table 2). carboxy-terminal contains groups which can be
Various further natural and synthetic opioid phosphorylated and which are involved in recep-
peptides, acting as agonists or antagonist at the tor internalization and inactivation. The seven
different opioid receptors have been discovered. transmembrane regions are connected by extra-
They have the same effect and side effect profile cellular and intracellular loop regions of differ-
as their nonpeptidic counterparts. Opioid pep- ent length. Comparing the sequence of the µ-, δ-,
tides are only used experimentally and no com- and κ-receptor reveals that the highest degree
pound has reached clinical application. of similarity is located in the transmembrane
regions and in the intracellular loop, whereas
Molecular Pharmacology of Opioid Re- the external loops and both terminal regions are
ceptors. The first successfully cloned opioid re- more heterogeneous. The external loops and the
ceptor was the δ-opioid receptor of the mouse, terminal part are involved in the selection and
which was in parallel described by the groups binding of the ligands and contain the structural
of Kieffer [349] and Evans [350]. Both used elements, which determine the receptor selectiv-
neuroblastoma-glioma hybridoma cells which ity.
expressed a high density of δ-receptors in their
membrane. The isolated receptor protein con- Transduction Mechanisms of Opioid Re-
sisted of 372 amino acids and had a mo- ceptor Interaction. As described above, all
lecular mass of 40 644. The amino acid se- four receptor types belong to the group of G-
quence showed a partial overlap with the recep- protein-coupled receptors (GPCRs) [354]. Ag-
tors of somatostatin (37 %), angiotensin (31 %) onistic binding at the receptor induces associ-
and interleukine-8 (22 %). Similar to the so- ation of the α-, β-, and γ- subunit of the G-
matostatin receptor, seven hydrophobic domains protein which triggers several biochemical reac-
were identified by which the receptor is in- tions within the cell [355–358]. Most important
serted into the lipid bilayer of the cell mem- for the pharmacological effects of the opioids
brane. Shortly thereafter the rat and human δ- are:
receptors were cloned. With the same technique
Analgesics and Antipyretics 45
Table 2. Mammalian endogenous opioid peptides
Precursor Endogenous peptide Amino acid sequence
Pro-opiomelanocortin β-endorphin YGGFMTSEKSQTPLVTLFKNAIIKNAYKKGE

Pro-enkephalin [MET]enkephalin YGGFM


[LEU]enkephalin YGGFL

Pro-dynorphin dynorphin A YGGFLRRIRPKLKWDNQ


dynorphin B YGGFLRRQFKVVT
α-neoendorphin YGGFLRKYPK
β-neoendorphin YGGFLRKYP

Pro-nociceptin/OFQ a Nociceptin FGGFTGARKSARKLANQ


a
OFQ = orphanin FQ.

• Activation of a hyperpolarizing K+ channel explains why opioids, in addition to their promi-


(inward rectifying K+ channel) nent inhibitory actions, have some stimulant ef-
• Inactivation of voltage-dependent Ca2+ fects, too.
channels (N-, P- and R-type)
• Inhibition of adenylate cyclase Relation of the Biological Effect to the Opi-
A further relevant mechanism involved in the oid Receptor Type. The peptide structure of the
inhibition of synaptic transmission by opioids four different opioid receptors is very similar
is a direct impairment of the exocytotic release [348] and this is the reason why most of the opi-
of neurotransmitters, induced by stabilization of oid receptor ligands show affinity for more than
the presynaptic membrane. one type of receptor. In most cases, one type is
Additional actions with only partially un- preferred and the action and side effect profile is
derstood relevance are activation of phospholi- dominated by the properties of this receptor. A
pases (PLH2, PLC7), activation of MAP kinases huge chemical effort was necessary to develop
and activation of some voltage dependent Ca2+ receptor-specific “selective“ ligands, but nowa-
channels (L-type and T-type). days several selective agonists and antagonists
As a result of these actions opioids inhibit for each type of opioid receptor exist (Table 3).
neurotransmission at the presynaptic and post- µ-Opioid Receptors. Binding experiments
synaptic site. The presynaptic inhibition mostly and investigations in pain models have shown
depends on the direct inhibitory effect on trans- that predominantly the µ-receptor and to a lesser
mitter exocytosis from membrane-associated degree the κ-and d-receptors mediate pain inhi-
storage vessels. This direct effect is increased by bition. Most of the clinically used opioid anal-
the inhibition of Ca2+ channels, since Ca2+ ions gesics [359] have prevalence for the µ-opioid
trigger the transmitter release. Activation of K+ receptor and this confirms that µ-receptor activa-
ions induces membrane hyperpolarization and tion is the common pain inhibiting mechanism of
this is the most important action component for these compounds. Since morphine is the proto-
postsynaptic inhibition. type, they are called “morphine-like”analgesics.
Activation of K+ channels, inactivation of Their side effect profile includes respiratory de-
2+ pression, constipation as well as addiction and
Ca channels and direct inhibition of neuro-
transmitter release are powerful mechanisms by dependence. All these effects, corresponding to
which opioids inhibit the neuronal transmission analgesia, are mediated via µ-opioid receptor
of the pain signal. activation. This is the reason why the primary
Opioid receptors are not only located at ex- goal of the development of synthetic opioids, the
citatory synapses, but are likewise expressed at separation of analgesia from the addiction and
inhibitory neurons. At these synapses, opioids dependence potential, has never been achieved.
inhibit the transmission of the inhibitory signal, The involvement of the same receptor popu-
and this inhibition of inhibition as the result, lation in analgesia and side effect profile of
induces excitation and an increased release of morphine and morphine-like opioids was im-
neurotransmitters in the innervated neuron.This pressively confirmed in µ-knockout mice [360,
46 Analgesics and Antipyretics
Table 3. Opioid receptor specifity
Receptor type Selective ligands Nonselective ligands
Agonist Antagonist Agonist Antagonist
MOR morphine CTOP levorphanol naloxone
µ- fentanyl etorphine naltrexone
methadone
DAMGO

KOR enadoline nor-BNI levorphanol naloxone


κ- U50,488 etorphine naltrexone
dynorphin

DOR DPDPE naltrindol levorphanol naloxone


δ- SNC-80 etorphine naltrexone
MOR = mu-opioid receptor. KOR = kappa-opioid receptor. DOR = delta-opioid receptor. DAMGO = [d-Ala2 , MePhe4 ,
Gly(ol)5 ]enkephalin. Enadoline = (5R)-(5α,7α,8β-(-)-N-methyl-N-(7-[1-pyrrolidinyl]-1-oxaspiro[4,5]dec-8-yl)-4-benzofuranacetamide,
previously CI977. U50,488 = trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolidinyl)-cyclohexyl]-benzeneacetamide methanesulfonate. DPDPE
= [d-Pen2 , d-Pen5 ]enkephalin. SNC-80 =
(+)-4-[(αR)-α-((2S,5R)-4-allyl-2,5-dimethyl-1-piperazinyl)-3-methoxybenzyl]-N,N-diethylbenzamide. CTOP =
d-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 . nor-BNI = nor-binaltorphimine.

361]. In these animals, morphine did neither δ-Opioid Receptors. In contrast to µ- and κ-
show an analgesic effect nor side effects. There receptors, indications for a prominent role of δ-
was no respiratory depression and no inhibi- receptors in the pain process are less obvious
tion of gastrointestinal motility and secretion. [364, 365]. There is a broad but still controversial
In behavioral models in µ-knockouts, morphine discussion concerning a genuine analgesic effect
did not induce liking or any other signs of ad- mediated by δ-1 or δ-2 receptors, because anal-
diction and repeated treatment with morphine gesia is scarcely observed in compounds with
did not produce signs of tolerance and physical pure δ-agonistic activity.
dependence. ORL-1 Receptors. The ORL-1 receptor [352,
κ-Opioid Receptors. Activation of κ- 366] differs in its peptide sequence from the clas-
receptors induces a clinically relevant pain in- sical opioid receptors. In contrast to these, ORL-
hibition [362, 363], which seems to be less 1 receptor activation at least at the supraspinal
efficacious than µ-receptor mediated analge- level has a pronociceptive effect and induces
sia. κ-Receptor interaction is a common action pain. Supraspinally induced release of spinal
component of the so-called partial opioid ago- CCK-8, NMDA or PGE-2 is discussed as the
nists or agonists – antagonists, such as penta- mechanism of pronociceptive activity. Spinal
zocine, nalbuphine, and butorphanol (see Sec- application of nociceptine incontrary induces
tion 3.1.1.1). The most prominent side effects of anti-nociception. With the aid of nonpeptidic
κ-activation are sedation and diuresis. In con- ORL-1 agonists and antagonists, which are cur-
trast to µ-agonists, which induce well-being and rently under development, it will be possible to
euphoria, activation of κ-receptors in the limbic elucidate the contribution of the ORL-1 opioid
system induces dysphoria and other unpleasant system to pain processing and to other physio-
psychic effects, such as hallucinations and spa- logical processes [367].
tial disorientation. The dose range of analgesia
and psychic side effects partly overlap and this Location of Opioid Receptors in the Pain
interferes with the medical use of κ-receptor ag- Pathway. The endogenous opioid system [368]
onists. In compounds with a marked κ-agonistic is the most important component of the pain in-
component (cyclazocine, nalorphine) the dys- hibitory system of the body. Opioids act at dif-
phoric side effects are so prominent that the ferent levels of the pain pathway and their action
compounds could not be used for therapeu- interferes with different aspects of the pain pro-
tic purposes. This may explain why despite an cessing [369, 370].
intensive search for κ-selective analgesics, no Opioids induce:
selective κ-agonist has ever reached the clinical • Inhibition of the transmission of the pain sig-
use. nal
Analgesics and Antipyretics 47

• Inhibition of the emotional aspect of pain clearly shown in well-controlled clinical stud-
• Inhibition of pain realization ies [372]. The most important use of opioids
Pain processing is inhibited at the spinal and in acute pain treatment is postoperative pain,
supraspinal level. Spinal opioid receptors are lo- whereas treatment of cancer pain, often accom-
cated pre- and postsynaptically at interneurons panied by a neuropathic pain component, is the
of the substantia gelatinosa of the dorsal horn. classical domain of chronic opioid treatment.
The opioid interneurons inhibit the release of
excitatory transmitters and reduce the transmis- Opioid Side Effects.
sion of the pain signal from the primary afferents Respiratory Depression. Opioids induce
to the secondary neurons of the spinal ascending respiratory depression via inhibition of the res-
pain pathway. Supraspinally, opioids are located piratory center of the medulla oblongata, which
in different regions of the brainstem, in the peri- respond to the pCO2 content of the blood [373,
aquaeductal grey matter, in the limbic system, in 374]. The inhibitory effect is more prominent in
the thalamic nuclei, in the basal ganglia, and in respect to the respiratory frequency than to the
the cortex. Cortical and thalamic localization is volume of respiration. At higher opioid dosages,
involved in the perception of the pain stimulus, respiration becomes irregular and grasping oc-
cortical regions in addition in the localization of curs. In awake persons, respiratory depression
the origin of the pain focus. by opioids can be voluntarily compensated over
Opioids in the different parts of the limbic a broad dose range. Respiratory impairment is
system suppress the emotional component of not a prominent feature in awake pain patients,
pain and the pain suffering. In the formatio retic- because pain itself is a strong stimulus for res-
ularis, they inhibit the pain induced activation of piration.
autonomous functions, such as increase in respi- Opioid-induced respiratory depression is
ration, increase in blood pressure and sweating. augmented by other CNS-depressant com-
Inhibitory actions at the formatio reticularis are pounds such as sedatives and hypnotics. Res-
the origin of the most prominent side effects of piratory depression becomes an important side
the opioids, like respiratory depression, brady- effect when opioids are used for postoperative
cardia and the centrally mediated part of the gas- pain treatment, because the anesthetic agent and
trointestinal inhibition. most adjuncts of anesthesia induce a long last-
In addition to the inhibitory effect at the as- ing depressant effect on respiration, which can
cending pain transmission, opioids activate a increase the opioid effects up to respiratory ar-
descending pain inhibitory system, which orig- rest. Therefore a careful supervision of respira-
inates from different centers of the pons and tion during the postoperative period is manda-
medulla, such as nucleus coeruleus, areas of the tory [375]. Opioid-induced respiratory depres-
periaquaeductal grey matter, and areas of the sion can be interrupted by the opioid antagonist
raphe nuclei. The descending nerve fibers termi- naloxone. Naloxone has a short duration of ac-
nate at the spinal interneurons in noradrenergic tion and repeated applications may be necessary
and serotoninergic inhibitory synapses, which to successfully counteract the effect of longer
suppress the ascending pain signal. Thus, opi- acting opioids. Highly potent opioids induce se-
oids inhibit the spinal pain processing by two vere respiratory depression in the higher dose
mechanisms, one is a direct pre- and postsynap- range. In addition, they induce a stiffness of the
tic inhibition of the ascending pain pathway and chest musculature [376], called chest rigidity or
the other is a centrally-mediated activation of the “wooden chest”, which is mediated via stimula-
descending pain inhibitory system. tion of dopamine release in the nucleus cauda-
tus. Chest rigidity further increases the respira-
Use of Opioids in Pain Treatment. Opioids tion impairment by these compounds. Therefore
are used for the treatment of moderate to se- higher doses of potent opioids such as fentanyl
vere or very severe pain of acute or chronic type and analogues, as used for anesthesia, need arti-
[371]. Nearly all forms of pain are sensitive to ficial or at least assisted ventilation.
opioid treatment and in contrast to traditional Cardiovascular Effects. Nearly all opioids
opinions even neuropathic pain is reasonably induce bradycardia [377], most likely mediated
sensitive to higher doses of opioids. This was
48 Analgesics and Antipyretics

via central stimulation of the vagus nerve. The Emetic Activity. Nausea and emesis are com-
cardiovascular depression of most opioids is mon unpleasant side effects of opioids [382,
moderate and only the stronger opioids of the 383]. They are most intensively experienced at
fentanyl group induce a more prominent effect. the beginning of the treatment. During chronic
Morphine and some analogues induce a non- application, tolerance may occur, which reduces
opioid receptor mediated release of histamine, the emetic sequelae. Nausea and emesis are in-
which can result in a decrease in blood pressure duced via activation of chemoreceptors, which
and a compensatory tachycardia. are located in the trigger zone of the area
Gastrointestinal Effects. Opioids induce an postrema of the formatio reticularis. The recep-
inhibitory effect on gastrointestinal motility and tors are located at the tissue surface and contact
fluid secretion [378]. The effect is peripherally the circulating blood. Thus the emetic effect of
and centrally mediated. The peripheral part is opioids is not mediated centrally, which means
related to µ- and κ-receptors at intestinal or- after penetration of the blood – brain barrier, but
gans, which are densely equipped with opioid rather peripherally via the part of the compound,
receptors. The receptors are located at parasym- which is distributed in the circulating blood.
pathic ganglions and inhibit the release of acetyl- After passage through the blood – brain bar-
choline, which stimulates the contraction of the rier, opioids have an antiemetic effect [384]. The
smooth muscles. Inhibition of the intestinal fluid emesis inhibition is induced via blockade of an
secretion is mediated via inhibition of adenylate emesis center located in a more central area
cyclase. The intestinal effects of opioids extend of the formatio reticularis. This explains why
to all parts of the gut and results in inhibition the emetic effect of opioids is most prominent
of stomach emptying and inhibition of secretion immediately after application, especially after
and motility in the duodenum, jejunum, colon, rapid intravenous administration and is reduced
and rectum. or terminated when the compound has reached
Reduced motility and secretion can let to con- the CNS. The more hydrophilic opioids such as
stipation, which is the most prominent side ef- morphine have stronger emetic side effects than
fect of chronic opioid treatment [379]. Opioid- lipophilic compounds like methadone or fen-
induced constipation can increase up to mega- tanyl [385], which are rapidly transported into
colon or paralytic ileus. Therefore chronic opi- the CNS.
oid treatment should be accompanied by con- Tolerance and Dependence. µ-Opioid com-
comitant use of laxatives. Besides their periph- pounds induce a feeling of well being and eu-
eral actions, opioids are involved in the central phoria, which is mediated by the release of
regulation of intestinal functions which is lo- dopamine within the limbic system. κ-Opioids
cated in the formatio reticularis. This explains induce an opposite effect with dysphoria, dis-
why the intestinal side effects of opioids are not orientation, and hallucinations [386]. Repeated
restricted to the more hydrophilic compounds activation of the µ-opioid rewarding system
such as morphine, but are seen likewise in the may induce a psychological dependence, which
more centrally active lipophilic analogues. Dur- leads to addiction and a compulsory behavior of
ing chronic opioid treatment a varying degree of drug seeking [387]. In addition, higher opioid
tolerance towards the intestinal side effects may dosages, as used for nonmedicinal purposes, in-
occur. duces tolerance and as a consequence a further
The intestinal inhibitory action of opioids can increase of the dose to achieve the intended ef-
be used for the treatment of diarrhea [380]. The fect. In the course of the tolerance development
clinically most important antidiarrheal opioid is opioid users becomes physically dependent on
loperamide [381]. After oral administration, lop- the supply of the compound [388] and suspen-
eramide acts locally within the gastrointestinal sion of the treatment or blocking of the opi-
tract. After parenteral administration, the com- oid receptors with an antagonist induces a with-
pound is rapidly inactivated and does not reach drawal reaction, characterized by strong dyspho-
the CNS. Therefore loperamide does not show ria, restlessness, pain, and various symptoms of
the typical central opioid side effects, has no autonomic dysregulation, like diarrhea, shiver-
analgesic action and has no abuse potential. ing, chills and cardiovascular collapse.
Analgesics and Antipyretics 49

The euphorigenic effect of opioids, the with water, precipitation with aqueous Na2 CO3 -
“opioid kick”, is more intensely induced by solution, washing of the precipitate with ethanol
lipophilic compounds such as diacetylmorphine and dissolving in diluted acetic acid [392–395].
(heroin), which rapidly penetrates into the CNS.
The feeling of euphoria on the one hand and the
absence of well-being on the other hand is more
prominent when brain concentrations of the opi-
oid change rapidly. This intensifies drug seeking
behavior and psychological as well as physical
dependence.
In contrast to recreational use, chronic pain
treatment with opioids afforts constant dosing Morphine in the conventional presentation
and has only a limited risk to induce psycho- (left) and in a stereoformula according to IU-
logical dependence and drug addiction [389]. A PAC rules (right)
proper dosing during chronic pain treatment can The total synthesis of (-)-morphine had been a
often postpone tolerance development for longer challenging target for organic chemists for many
time periods. The most important precaution to decades [316, 334, 396]. Although a number of
avoid tolerance and dependence development is successful syntheses were developed, only a few
to ensure constant plasma levels of the opioid, could produce the compound in an enantio- and
which have to be high enough to give complete diastereo-controlled manner [397–399].
pain relief. This can be reasonably achieved by Morphine biosynthesis: The studies on the
oral administration of retarded formulations or biosynthesis of morphine have been performed
by an opioid patch [390]. Breakthrough pain, on cell cultures mainly of Coptis japonica and
which is often induced by a fluctuation in pain species of Thalictrum. The overall biosynthetic
intensity, should be rapidly compensated by ad- pathway from tyrosine to morphine is depicted
ditional treatment with an immediate release for- in Figure 3.
mulation of the same or a similar opioid. Opioid receptor binding: Morphine has a
high (nanomolar) binding affinity for the µ-
opioid receptor. The affinity for the δ- and κ-
3.1.1. Opioids in Clinical Use opioid receptor is at least 10-fold lower.
Clinical use: Morphine is a very potent anal-
3.1.1.1. Morphine and Morphinane gesic and still the “gold standard” for the treat-
Derivatives ment of severe, acute, and chronic pain [400].
The compound is used in various salt forms,
Morphine [57-27-2], (5α,6α)-7,8-di- preferentially as the hydrochloride or sulfate.
dehydro-4,5-epoxy-17-methylmorphinan-3,6- Morphine can be administered orally or par-
diol, C17 H19 NO3 , M r 285.34, mp 254 ◦ C (de- enterally. Oral administration is preferred for
comp., 197 ◦ C is reported for a metastable chronic pain treatment and various slow-release
25
phase), [α]D −132◦ (c = 1, CH3 OH); hy- forms have been developed to reduce the ap-
drochloride [52-26-6], C17 H19 NO3 · HCl, plication frequency to 1–2 times per day. Par-
25
M r 321.80, mp 200 ◦ C (trihydrate), [α]D enterally, morphine is used in doses of 10

−113.5 (c = 2.2, water); sulfate (2 : 1) [64- mg mostly for postoperative pain and self-
31-3], C17 H19 NO3 · 1/2 H2 SO4 , M r 668.76; administration devices have been developed for
sulfate pentahydrate [6211-15-0], C17 H19 NO3 · patient-controlled analgesia (PCA).
1/2 H2 SO4 · 5H2 O, M r 758.85, mp 250 ◦ C (de- Other morphine preparations are under eval-
25
comp.), [α]D −108.7◦ (c = 4, water). Mor- uation for local administration, e.g., inhalation
phine is the main alkaloid of opium, isolated via the broncho-pulmonal way.
from the milky liquid of the poppy seed capsule Pharmacokinetic properties: Morphine is ex-
[108]. tensively metabolized by glucuronidation at
Preparation: Morphine is obtained by extrac- both hydroxyl groups at positions 3 and 6 and by
tion of poppy capsules or opium (opium contains N-demethylation [401, 402]. The 3-glucuronide
9–14 % morphine, depending on the source) has a minimal opioid receptor binding affinity
50 Analgesics and Antipyretics

Figure 3. The overall biosynthetic pathway from tyrosine to morphine


Analgesics and Antipyretics 51

and is devoid of analgesic action, whereas the atives. Other frequent side effects are nausea,
morphine-6-glucuronide has a similar binding vomiting, dizziness, and sedation.
affinity as morphine, is analgesically active and Morphine is a controlled substance. It has a
seems to be involved in pain inhibition during high euphorigenic potential and is liable to abuse
chronic oral treatment with morphine [403]. De- and dependence. The euphorigenic effect is less
spite the polar sugar residue the glucuronide can expressed in the context of pain treatment and
pass the blood – brain-barrier. The morphine- tolerance and dependence can largely be avoided
6-glucuronide is being tested as an indepen- by appropriate dosing and application intervals,
dent opioid analgesic. The N-demethyl deriva- securing constant and pain-appropriate plasma
tive normorphine is analgesically active, but has levels of the compound.
a lower µ-receptor affinity, a lower potency and Trade names: M-Long (Germany), MST con-
a shorter duration of action than morphine. Nor- tinuous (Germany, UK), Capros (Germany), Ka-
morphine is not in clinical use. panol (Germany, USA), Moscontin (France), In-
The main metabolites of morphine are shown fumorph (USA), Oxamorph (USA).
in Figure 4. Morphine is also marketed in many generic
formulations.

Codeine [76-57-3], C18 H21 NO3 , M r


299.36, mp 154–156 ◦ C (monohydrate from wa-
ter or diluted alcohol), anhydrous form sublimes
25
at 140–145 ◦ C and 133.32 Pa, [α]D −136◦
(c = 2, CH3 CH2 OH); monohydrate [6059-47-
8], C18 H21 NO3 · H2 O, M r 317.38, mp 154–
156 ◦ C; hydrochloride dihydrate C18 H21 NO3 ·
HCl · 2 H2 O, M r 371.86, mp 280 ◦ C (decomp.),
[α]D − 108◦ ; hydrobromide dihydrate [125-25-
7], C18 H21 NO3 · HBr · 2 H2 O, M r 416.31, mp
190–192 ◦ C (anhydrous), [α]D − 96.6◦ ; phos-
phate [52-28-8].
Preparation: Codeine is extracted from
opium (concentration in opium 0.7 to 2.5 %, de-
pending on the source), but mostly prepared by
methylation of morphine in a phase transfer re-
action [404, 405].

Figure 4. Metabolic pathway of morphine

Side effects: Morphine induces a variety of


centrally and peripherally mediated side effects.
Most important after parenteral administration
is respiratory depression, especially in the post-
operative situation. Chronic oral administration
induces constipation and chronic treatment with
oral morphine has to be supplemented with lax-
52 Analgesics and Antipyretics

Opioid receptor binding: Codeine has a low


affinity at µ-, δ-, and κ-opioid receptors and the
in vivo effects are predominantly induced by me-
tabolization to morphine [55].
Clinical use: Codeine [407] is an O-methyl
analogue of morphine. It has a morphine-like
action profile but a 5–10 times lower potency.
Codeine has a good oral availability and oral
administration forms are used for the treatment
of mild to moderate pain and as antitussive (→
Cough Remedies, Section 2.1.1). In therapeu-
tic dosages, codeine produces fewer opioid-type
side effects than morphine and has only a low
risk of abuse. The lower abuse and dependence
can be explained by the fact that the opioid ac-
tion is only available after metabolic activation. Clinical use: Ethylmorphine (→ Cough
High codeine doses can induce excitement and Remedies, Section 2.1.1) is the ethyl congener
convulsions. of codeine and has a similar therapeutic applica-
The compound is extensively metabolized tion. It has a low opioid receptor affinity and is
by O- and N-demethylation followed by glu- metabolized to the active metabolites morphine
curonidation. The main metabolites are nor- and normorphine. Ethylmorphine can be used
codeine, morphine, and hydrocodeine and their as cough suppressant, medium-potent analgesic,
glucuronides. There are indications [408] that and anti-diarrheal agent. Today the compound is
the analgesic effect is reduced in poor metabo- widely out of therapeutic use, but is often used
lizers with a low CYP2D6 activity. as clinical reagent to characterize the metabolic
Codeine is used in the form of different salts N-dealkylation activity of CYP3A4 [411, 412].
like hydrochloride, phosphate and sulfate. To in- Trade names: Codethyline (Belgium), Tra-
crease the duration of action, slow-release prepa- chyl (France), Collins Elixir (UK).
rations have been developed. Codeine salts are
frequently combined with non-opioid analgesics Diacetylmorphine [561-27-2], diamor-
such as paracetamol, acetylsalicylic acid, or di- phine, heroin, C21 H23 NO5 , M r 369.41, mp
clofenac. 25
173 ◦ C, [α]D −166◦ (c = 1.49, CH3 OH);
Trade names: Codipront (Germany), Codi-
hydrochloride monohydrate [561-27-2],
caps (Germany), Codimal (USA).
C21 H23 NO5 · HCl · H2 O, M r 423.89, mp 243–
Combination with paracetamol: Co-Tylenol 25
(USA). 244 ◦ C, [α]D −156◦ (c = 1.044).
Combination with Diclofenac: Combaren Synthesis: Diamorphine is prepared by acety-
(USA). lation of morphine with acetic anhydride [413].
Codeine is also marketed in many generic for-
mulations.

Ethylmorphine [76-58-4], (5α,6α)-


7,8-didehydro-4,5-epoxy-3-ethoxy-17-methyl-
morphinan-6-ol, C19 H23 NO3 , M r 313.40, mp
199–201 ◦ C; hydrochloride dihydrate [6746-
59-4], C19 H23 NO3 · HCl · H2 O M r 385.89, mp
123 ◦ C (decomp.), anhydrous form melts at
170 ◦ C (decomp.). Opioid receptor binding: Diamorphine [414]
Synthesis: Ethylmorphine is synthesized by has a 10- to 100-fold lower µ-opioid receptor
ethylation of morphine with ethyl benzenesul- binding affinity than morphine. The relevant opi-
fonate [48, 409, 410]. oid properties originate from the high µ-receptor
Analgesics and Antipyretics 53

affinity of the metabolites 6-acetylmorphine and [421] and as antitussive [422] (→ Cough Reme-
morphine [415]. dies, Section 2.1.1). Dihydrocodeine is mostly
Clinical use: Diamorphine [416] is a used as oral immediate- or sustained-release for-
lipophilic morphine derivative which rapidly mulations [423]. For pain treatment the dose
penetrates into the central nervous system, range is 30–80 mg, for cough inhibition doses
where 6-acetylmorphine and morphine are re- are in the range of 10 mg.
leased [417, 418]. The rapid brain access induces Dihydrocodeine induces morphine-like side
an immediate onset of action and this seems to be effects, but the intensity is less pronounced.
the reason for the strong euphorigenic effect and Chronic treatment may produce dependence,
the high abuse potential. Because of the wide- and abuse by opioid addicts has been reported.
spread abuse, therapeutic application is prohib- Trade names: Paracodin (Germany), Di-
ited in many countries including Germany and codin (France), DHC Continus (UK), Synalgos
the US. In other countries like the UK, parenteral (USA).
heroin is used for the relief of severe pain, espe-
cially in terminal illness. The compound is more Etorphine [14521-96-1], [5α,7α(R)]-4,5-
potent than morphine, but has a similar action epoxy-3-hydroxy-6-methoxy-α,17-dimethyl-
and opioid side effect profile. α-propyl-6,14-ethenomorphinan-7-methanol,
Trade name: Diagesil (UK). C25 H33 NO4 , M r 411.53, mp 214–217 ◦ C; hy-
drochloride [13764-49-3], C25 H33 NO4 · HCl,
Dihydrocodeine [125-28-0], (5α,6α)- M r 447.99, mp 266–267 ◦ C.
4,5-epoxy-3-methoxy-17-methylmorphinan-6- Synthesis: Starting from thebaine etorphine
ol, C18 H23 NO3 , M r 301.38, mp 112–113 ◦ C; can be synthesized in a similar way as buprenor-
tartrate (1 : 1) [5965-13-9], C18 H23 NO3 · phine (see page 66) [424–427].
C4 H6 O6 , M r 451.47, mp 192–193 ◦ C (com-
mercial medicinal grade usually melts at 186–
25
190 ◦ C, [α]D −72◦ to −75◦ (c = 1.0, water).
Synthesis: Hydrogenation of codeine yields
dihydrocodeine [48, 419, 420].

Opioid receptor binding: Etorphine [428] has


a high affinity and selectivity for the µ-opioid re-
ceptor.
Clinical use: Etorphine [429] is an extremely
strong and rapid-acting µ-opioid with a potency
400–1000-fold higher than morphine. The com-
pound is very rapidly absorbed through skin and
mucosa and extraordinary care is necessary to
avoid contamination during medical use. Nalox-
one or diprenorphine can be used as antago-
nist. Etorphine induces strong central nervous
Opioid receptor binding: Dihydrocodeine depression and is mostly used in veterinary prac-
has a low µ-opioid receptor binding affin- tice for immobilization or pain treatment of big
ity and opioid properties are mostly due to animals [430]. Etorphine is only used parenter-
metabolic activation to the demethyl derivative ally.
dihydromorphine. Trade name: Immobilon (UK).
Clinical use: Dihydrocodeine has codeine-
like analgesic and antitussive properties and is Hydrocodone [76-42-6], (5α)-4,5-
used for the treatment of moderate to severe pain epoxy-3-methoxy-17-methylmorphinan-6-one,
54 Analgesics and Antipyretics

C18 H21 NO3 , M r 299.36, mp 198 ◦ C; hy- Hydromorphone [466-99-9], (5α)-4,5-


drochloride [25968-91-6], C18 H21 NO3 · HCl, epoxy-3-hydroxy-17-methylmorphinan-6-one,
M r 335.83; hydrochloride monohydrate [124- C17 H19 NO3 , M r 285.34, mp 266–267 ◦ C;
90-3], C18 H21 NO3 · HCl · H2 O, M r 353.85, monohydrochloride [71-68-1], C17 H19 NO3 ·
27
mp 185–186 ◦ C (decomp.), [α]D −130◦ (c = HCl, M r 321.80.
2.877); bitartrate hemipentahydrate [34195-34- Synthesis [48, 435, 436]:
1], C18 H21 NO3 · C4 H6 O6 · 5/2 H2 O, M r 494.5, Morphine is hydrogenated over a palladium
mp 118–128 ◦ C. catalyst, and the resulting dihydromorphine is
Synthesis: Palladium or platinum catalyzed oxidized with benzophenone and potassium
isomerization of codeine yields hydrocodone tert-butoxide. Alternative oxidants are cyclo-
[48, 431, 432]. hexanone with aluminum tri(tert-butoxide) or
aluminum triphenoxide.

Opioid receptor binding: Hydromorphone


has a high affinity and selectivity for the µ-opioid
receptor, the µ-affinity is about 10-fold higher as
compared to morphine.
Clinical use: Hydromorphone [437] is a
morphine derivative with an analgesic potency
higher than morphine. It is used as hydrochloride
Opioid receptor binding: Hydrocodone has a parenterally or orally in the dose range of 2–4
µ-opioid receptor binding affinity that is 10- to mg to treat moderate to severe pain [438] or as
100-fold higher than that of codeine [433]. In a syrup for cough inhibition. Hydromorphone
contrast to codeine, the compound itself has a shows the typical morphine-like side effect pro-
relevant µ-opioid receptor affinity and is, in ad- file and has a relatively high potential for addic-
dition to the active metabolite hydromorphone, tion and dependence [439].
responsible for analgesia and other opioid prop- Trade name: Dilaudid (Germany, UK, USA),
erties. Palladone (USA).
Clinical use: Hydrocodone [420] is an opioid
related to codeine with analgesic and marked an- Levorphanol [77-07-6], 17-methylmorphi-
titussive properties. The compound is more po- nan-3-ol, (−)-3-hydroxy-N-methylmorphinan,
tent than codeine and is used in oral doses of 5 C17 H23 NO, M r 425.48, mp 198–199 ◦ C,
20
to 10 mg for the treatment of moderate to mod- [α]D −56◦ (c = 3, CH3 CH2 OH); tartrate di-
erately severe pain. Combinations with parac- hydrate [5985-38-6], C17 H23 NO · C4 H6 O6 · 2
etamol or other COX inhibitors are common, as H2 O, M r 257.37, mp 113–115 ◦ C (anhydrous
well as multi-ingredient preparations as antitus- 20
mp 206–208 ◦ C), [α]D −14◦ (c = 3, water).
sives. Hydrocodone induces side effects similar Synthesis: The analgesic activity of racemor-
to codeine and morphine and has an appreciable phan is due to the (−) isomer, levorphanol,
abuse and dependence potential [434]. which is obtained by resolving the racemate
Trade names: Dicodid (Germany, UK), Vi- with (+)-d-tartaric acid. The resolution can also
codin (USA). be performed on the intermediate 1-(4-methoxy
Combination with paracetamol: Zydone benzyl)-1,2,3,4,5,6,7,8-octahydro isoquinoline
(USA), combination with ibuprofen: Vicopro- (1) prior to N-methylation [48, 440–443].
fen (USA).
Analgesics and Antipyretics 55

The compound induces morphine-type addic-


tion and dependence [446].
Trade name: Levo-Dromoran (USA).

Oxycodone [76-42-6], (5α)-4,5-epoxy-14-


hydroxy-3-methoxy-17-methylmorphinan-6-
one, 14-hydroxydihydrocodeinone, dihydrone,
dihydrohydroxycodeinone, C18 H21 NO4 , M r
315.36, mp 218-220 ◦ C; hydrochloride [124-
90-3], C18 H21 NO4 · HCl, M r 351.81, mp 270–
20
272 ◦ C (decomp.), [α]D −125◦ (c = 2.5, water).
Synthesis [48, 447, 448]: Thebaine is ox-
idized with hydrogene peroxide to 14-hy-
droxycodeinone [449, 450], which is hydro-
genated directly or via its oxime, or its bromi-
nation products to oxycodone. The reduction
of 14-hydroxycodeinone can also be performed
with sodium hydrosulfite. Alternatively, 14-hy-
droxycodeinone is prepared by oxidation of
codeine (see next page, top).
Opioid receptor binding: Oxycodone [433] is
a µ-selective opioid with a 10-fold higher recep-
tor affinity than codeine. Both the parent com-
pound and the high affinity metabolite oxymor-
phone mediate the opioid effects of the com-
pound [451].
Clinical use: Oxycodone [452] is an orally
active opioid analgesic for the treatment of mod-
erate to moderately severe pain. The compound
is used as hydrochloride in a dose range of 5–
10 mg and is often combined with paracetamol
or other COX-inhibitors. Higher dosages and
oral slow-release preparations [453] are used
for the control of severe pain. Oxycodone has
a morphine-like side effect profile. Respiratory
depression has been found in children. The com-
pound has a relevant abuse and dependence po-
tential and illicit use of the retarded prepara-
tions is reported. To obviate misuse, abuse deter-
rent formulations (ADF) and combinations with
naloxone and naltrexone are under development.
Opioid receptor binding: Levorphanol is a µ-
Trade names: Eukodal (Germany), Eubine
selective synthetic opioid with a higher receptor
(France), Proladone (UK),Roxicodone, Oxy-
affinity than morphine [225].
Contine (USA), Oxygesic (Germany)
Clinical use: Levorphanol [445] is a strong
Combination with ibuprofene: Combunox
analgesic used as tartrate for parenteral and oral
(USA).
administration. Parenterally, levorphanol is 4–
5 times more potent than morphine. After oral Oxymorphone [76-41-5], (5α)-4,5-
administration, the compound has a long du- epoxy-3,14-hydroxy-17-methylmorphinan-
ration of action (up to 8 h). Levorphanol has a 6-one, dihydrohydroxymorphinone, di-
morphine-type side effect profile with prominent hydro-14-hydroxymorphinone, 14-hydroxydi-
respiratory depression in the high dose range. hydromorphinone, C17 H19 NO4 , M r 301.34, mp
56 Analgesics and Antipyretics

248–249 ◦ C (decomp.); hydrochloride [357-07- 3.1.1.2. Piperidine Derivatives


3], C17 H19 NO4 · HCl, M r 337.80.
Synthesis [454] Oxycodone is hydrolyzed 3.1.1.2.1. Meperidine and Congeners
with boiling concentrated hydrobromic acid.
(for Diphenoxylate, Loperamide, and Pir-
itramide see Section 3.1.1.4)

Pethidine [57-42-1], meperidine, ethyl


1-methyl-4-phenylpiperidine-4-carboxylate,
C15 H21 NO2 , M r 247.33; hydrochloride [50-
13-5], C15 H21 NO2 · HCl, M r 283.80, mp 186–
189 ◦ C.

Opioid receptor binding: Oxymorphone is a


µ-selective opioid with a binding affinity in the
range of morphine.
Clinical use: Oxymorphone is a synthetic
morphine derivative with a potency 5–10 times
higher than that of morphine. The compound
is used as hydrochloride in parenteral and rec-
tal application forms (suppositories) and as oral
sustained-release formulations to treat moder-
ate to severe pain [455]. Oxymorphone has no
cough suppressant activity. Oxymorphone has a
morphine-type side effect profile and can induce
addiction and dependence [456].
Trade name: Numorphan (USA, Canada).
Analgesics and Antipyretics 57

Synthesis [48, 457–460]: dine [463]. Severe side effects including coma
a) The original synthesis involved condensation and cyanosis have been observed in combina-
of benzyl cyanide with N,N-bis(2-chloro- tion with MAO-inhibitors [464]. Pethidine in-
ethyl)-N-methyl-amine, which is a skin ir- duces morphine-type tolerance and dependence
ritant and a carcinogen. and addicts using high doses of pethidine have
b) Another synthesis begins with pyridine-4- an increased risk of excitatory side effects.
carboxylic acid. Due to the high risk of severe side effects the
clinical use of the compound has strongly de-
creased [465–467].
Trade names: Dolantin (Germany), Dolosal
(France), Demerol (USA).

Ketobemidone [469-79-4], N-(3-hy-


droxyphenyl)-N-(1-methylpiperidin-4-yl)-
propionamide, C15 H21 NO2 , M r 247.34,
mp 150–151 ◦ C; hydrochloride [5965-49-1],
C15 H21 NO2 · HCl, M r 283.80, mp 201–202 ◦ C.
Synthesis: The cyano group of 4-cyano-4-
(3-methoxyphenyl)-1-methylpiperidine is con-
verted into a ketoethyl group by reaction with
ethylmagnesium bromide. Subsequent ether
cleavage by means of HBr yields ketobemidone
[48, 459, 468, 469].

Opioid receptor binding: Pethidine is a µ-


selective synthetic opioid with an intermediate
receptor affinity [461].
Clinical use: Pethidine [462] is a synthetic
analgesic with µ-receptor preference similar to
morphine. Pethidine, introduced in 1939, was
the first fully synthetic analgesic with morphine-
like activity. It has a lower potency and a shorter
duration of action than morphine and is used as
hydrochloride for the treatment of moderate to
severe pain. It is preferentially used in parenteral
formulations, perioperative and obstetrical anal-
gesia and as adjunct to anesthesia.
Pethidine has a morphine-type side effect
profile with a lower incidence of constipation.
Higher doses induce central stimulation and an- Opioid receptor affinity: Ketobemidone is
timuscarinic effects accompanied by pupil di- a µ-selective synthetic opioid with a receptor
latation. In even larger doses toxic symptoms affinity similar to morphine [470].
such as muscular twitching, tremor, mental con- Clinical use: Ketobemidone is a synthetic
fusion, and convulsions occur, which are partly opioid of the pethidine group with an analgesic
attributed to the more toxic metabolite norpethi- potency in the range of morphine [471]. The
58 Analgesics and Antipyretics

compound has a weak NMDA blocking effect Fentanyl and fentanyl derivatives (so-called
[472], which may contribute to the analgesic ef- designer drugs) have an essential abuse poten-
ficacy. It can be given orally, by injection or rec- tial [477] and induce a morphine type of physical
tally; usual doses are 5–10 mg. Ketobemidone dependence.
has a morphine-like side effect and abuse and Trade names: Actic (USA), Duragesic
dependence potential [473]. (USA), Durogesic, Durogesic-SMAT (Ger-
Trade names: Cliradon (Germany, out of many), Durogesic-Dtrans (UK), Fentanyl
use), Ketogan (Sweden, Norway). Janssen (Germany), Sublimaze (UK, USA).
Combination with Droperidol: Thalamonal.
Transdermal fentanyl patches are also mar-
3.1.1.3. Fentanyl and Congeners keted in generic formulations.

Fentanyl [437-38-7], N-(1-phenethylpiperi-


din-4-yl)-N-phenylpropionamide, C22 H28 N2 O,
M r 336.47, mp 83–85 ◦ C; citrate (1 : 1) [990-
73-8], C22 H28 N2 O · C6 H8 O7 , M r 528.60, mp
149–151 ◦ C.
Synthesis [48, 474] (see right column).
Opioid receptor binding: Fentanyl is a µ-
selective potent opioid with a similar recep-
tor binding affinity as morphine. The higher in
vivo potency of fentanyl results from its higher
lipophilicity.
Clinical use: Fentanyl is a potent synthetic
opioid related to pethidine [475] and has an ac-
tion profile similar to morphine. Fentanyl citrate
is used for the inhibition of acute and chronic
severe pain, and especially as adjunct to anes-
thesia and as a primary anesthetic for induction
and maintenance of anesthesia [476]. In com-
bination with neuroleptics or tranquilizers it in-
duces a surgical state named neuroleptanalgesia
in which surgery can be performed in an awake,
but calm and pain-free status. Fentanyl is orally
bioavailable and can be administered orally (spe-
cial forms exist for the oral-transmucosal route),
parenterally, and transdermally as patch formu-
lations. A formerly used less safe reservoir patch
formulation has now been substituted by a new
matrix patch. Fentanyl is a highly lipophilic
compound and is bound to plasma proteins to
about 80 %. The compound has a rapid onset
and a relatively short duration of action, presum-
ably due to tissue redistribution. The elimination
half-life is longer than that of morphine.
The side effect profile is typical of potent µ-
opioids with respiratory depression, increased
muscle tone (chest wall rigidity during fentanyl
anesthesia), strong sedation, and emesis being
most prominent. Adverse reactions can be an-
tagonized with naloxone.
Analgesics and Antipyretics 59

Alfentanil [71195-58-9], N-{1-[2-(4-ethyl-


5-oxo-4,5-dihydrotetrazol-1-yl)ethyl]-4-meth-
oxymethylpiperidin-4-yl}-N-phenylpropion-
amide, C21 H32 N6 O3 , M r 416,25; hydrochloride
monohydrate [70879-28-6], C21 H32 N6 O3 ·
HCl · H2 O, M r 470.99, mp 138.4–140.8 ◦ C.
Synthesis: The cyclization of ethyl isocy-
anate with sodium azide in the presence of
AlCl3 in refluxing THF gives 1-ethyl-1,4-di- Finally the tetrazole derivative 2 is condensed
hydro-5H-tetrazol-5-one, which is alkylated with the propionamide 3 in the presence of
with 1-chloro-2-bromoethane using Na2 CO3 Na2 CO3 and KI as catalysts in refluxing 4-
and KI in refluxing 4-methyl-2-pentanone to af- methyl-2-pentanone.
ford 1-ethyl-4-(2-chloroethyl)1,4-dihydro-5H-
tetrazol-5-one (2) [48, 478–480].

N-(4-methoxymethyl-4-piperidinyl)-N-
Opioid receptor binding: Alfentanil is a µ-
phenylpropionamide (3) is synthesized ac-
selective opioid [481] with a receptor affinity in
cording the following scheme starting from
the range of morphine and fentanyl.
1-benzyl-4-piperidone:
Clinical use: Alfentanil is a potent opioid
analgesic with a rapid onset and a shorter du-
ration of action than fentanyl [482]. The i. v.
formulation (dose range 1–4 mg) is mainly used
perioperatively as strong analgesic, supplement
to general anesthesia, or as a primary anesthetic
[483]. Alfentanil has a strong respiratory de-
pressant action and high doses induce chest wall
rigidity. The compound has a µ-type addiction
and dependence potential.
Trade names: Rapifen (Germany, France,
UK), Alfenta (USA).

Remifentanil [132875-61-7], methyl 1-(2-


methoxycarbonylethyl)-4-(phenylpropionylami-
no)piperidine-4-carboxylate, methyl 4-(meth-
oxycarbonyl)-4-[(1-oxopropyl)phenylamino]-
1-piperidinepropanoate, C20 H28 N2 O5 , M r
376.45; monohydrochloride [132539-07-2],
C20 H28 N2 O5 · HCl, M r 412.91; oxalate (1 :
1) [132875-62-8], C20 H28 N2 O5 · C2 H2 O4 , M r
466.49.
60 Analgesics and Antipyretics

Synthesis: Remifentanil is prepared by con- Opioid receptor binding: Sufentanil is a µ-


densation of methyl 4-(phenylpropionylamino) selective opioid with an about 10-fold higher re-
piperidine-4-carboxylate with methyl acrylate in ceptor affinity than fentanyl [497].
hot acetonitrile [48, 484–486].

Clinical use: Remifentanil [487, 488] is a Clinical use: Sufentanil [498] is a very potent
very short-acting and potent µ-opioid agonist and short-acting fentanyl derivative with pref-
with strong analgesic and anesthetic properties. erence for the µ-opioid receptor. It has strong
It has been developed as an ultra-short anes- sedating and analgesic properties and is used
thetic and adjunct to general anesthesia. [489, predominantly as an adjunct in anesthesia or
490]. It affords potent intraoperative analgesia as a primary anesthetic. Anesthetic doses in-
and has a sparing effect on concomitantly used duce respiratory depression and chest wall rigid-
sedatives and hypnotics [491]. Remifentanil is ity [499] which requires assisted ventilation.
given as intravenous short infusion in doses of For pain treatment intravenous or epidural on-
0.5–1 µg/kg or as continuous infusions in the demand procedures are in use and a patch for-
range of 0.0025–2 µg kg−1 min−1 . It is rapidly mulation for pain relief up to seven days is under
inactivated by plasma and tissue esterases [492]. development. Following parenteral administra-
The terminal elimination half-life is 10–20 min. tion sufentanil has a rapid onset and a short dura-
Remifentanil has a µ-opioid type side effect tion of action. The compound is very lipophilic
profile with strong CNS and respiratory depres- and has a high plasma protein binding of ≈ 90 %.
sant properties and a morphine-like addiction The short duration of action is more dependent
and dependence potential. on redistribution than on metabolic inactivation.
Trade name: Ultiva (Germany, UK, USA). Doses up to 8 µg/kg are adequate for pain treat-
ment and higher doses up to 30 µg/kg for surgery
Sufentanil [56030-54-7], N-[4-(meth- [500]. Sufentanil has a morphine-type side ef-
oxymethyl)-1-[2-(2-thienyl)ethyl]-4-piperi- fect profile and induces severe respiratory de-
dinyl]-N-phenylpropanamide, N-[4-methoxy- pression and chest wall rigidity in anesthetic
methyl-1-(2-thiophen-2-yl-ethyl)piperidin-4- dosages [501]. High dose levels have been asso-
yl]-N-phenylpropionamide, C22 H30 N2 O3 S, M r ciated with seizures. Prolonged use may induce
386.56, mp 96.6 ◦ C; citrate (1 : 1) [60561-17-3], tolerance and dependence.
C22 H30 N2 O3 S · C6 H8 O7 , M r 578.68. Trade name: Sufenta (Belgium, Germany,
Synthesis: Sufentanil is obtained by con- France, Norway, USA).
densation of N-(4-methoxymethylpiperidin-4-
yl)-N-phenylpropionamide with 2-thiophen-2-
3.1.1.4. Methadone and Congeners
yl-ethyl methanesulfonate [48, 493–496].
Levomethadone [125-58-6], (R)-6-
dimethylamino-4,4-diphenylheptan-3-one,
Analgesics and Antipyretics 61

C21 H27 NO, M r 309.45, mp 98–100 ◦ C (mp dence potential. Because of its slow elimination
20
racemic form [57-42-1] 78–79 ◦ C), [α]D −32◦ withdrawal reactions are more protracted and
(c = 1.8, CH3 CH2 OH), hydrochloride [5967- less severe than with morphine [517].
73-7], C21 H27 NO · HCl, M r 345.91, mp 240– Trade names: Eptadone (Italy), Dolophine
241 ◦ C (mp racemic form [1095-90-5] 231 ◦ C), (USA), Metasedin (Spain),Physeptone (UK), l-
20
[α]D −169◦ (c = 2.0, water). Polamidon (Germany).
Synthesis [502–509]:
Levomethadyl acetate [34433-66-4],
l-α-acetylmethadol (LAAM), 4-dimethyl-
amino-1-ethyl-2,2-diphenylpentyl acetate,
[S-(R*,R*)]-β-[2-(dimethylamino)propyl]-
α-ethyl-β-phenylbenzeneethanol acetate,
C23 H31 NO2 , M r 353.50; hydrochloride
C23 H31 NO2 · HCl, M r 389.96, mp 215 ◦ C,
25
[α]D −60◦ (c = 0.2, water).
Synthesis: Levomethadyl acetate is prepared
by reduction of dextromethadone and subse-
quent acylation [518].

Opioid receptor binding: Levomethadyl ac-


Opioid receptor binding: Levomethadone etate has a moderate affinity for opioid receptor
is the µ-selective levorotatory enantiomer of binding with selectivity for the µ-type. Higher
racemic methadone [510]. It has an opioid re- binding affinity is induced by the active metabo-
ceptor affinity in the range of morphine. lites l-alpha-noracetylmethadol, normethadol
Clinical use: Levomethadone [511] is similar and methadol [519].
to morphine in potency and side effect profile. Clinical use: Levomethadyl acetate is an
The compound has a high oral availability and a orally active methadone derivative with a slow
long duration of action [12, 513]. Methadone is onset and a long duration of action. The com-
extensively protein-bound and accumulation can pound is extensively metabolized and active
occur during chronic administration. For pain metabolites contribute to the long duration of
treatment, the active dose range is 2.5–10 mg. action. Levomethadyl acetate was used mainly
Methadone can be given orally or by subcuta- for substitution treatment of opioid dependence
neous or intramuscular administration (2.5–10 [520]. Oral doses from 20 up to 140 mg are given
mg in analgesia), but causes pain at the injection every 2–3 d. Supplementation with short acting
site. opioids is necessary during the first days because
The long duration of action makes the com- of the delayed onset of action.
pound suitable for the substitution treatment of Levomethadyl acetate induces opioid-type
opioid addiction [514]. Whereas levomethadone side effects with respiratory depression, brady-
is preferred for pain treatment, oral racemic cardia, and impairment of cardiac contractil-
methadone is widely used for the substitution ity [521]. The compound inhibits cardiac elec-
treatment of opioid addiction [515]. tric conductivity and increases the QT interval
Levomethadone has a morphine-like side ef- in the electrocardiogram [522] and may induce
fect profile with stronger respiratory depression ventricular arrhythmias and Torsade de pointes
and less sedation than morphine [516]. The com- [523]. Because of the cardiovascular side effects
pound has a morphine-type abuse and depen-
62 Analgesics and Antipyretics

(Q/T-interval prolongation), the compound was Dextropropoxyphene [469-62-5], 1-


recently withdrawn from the market. benzyl-3-dimethylamino-2-methyl-1-phenyl-
Trade name: Orlaam (USA). propionate, [S-(R*,S*)]-α-[2-(dimethylami-
no)-1-methylethyl]-α-phenylbenzeneethanol
Dextromoramide [357-56-2], (+)-(S)- propanoate (ester), C22 H29 NO2 , M r 339.47,
25
3-methyl-4-morpholin-4-yl-2,2-diphenyl-1- mp 75–76 ◦ C, [α]D +67.3◦ (c = 0.6, CHCl3 );
pyrrolidin-1-yl-butan-1-one, C25 H32 N2 O2 , M r hydrochloride [1639-60-7], C22 H29 NO2 · HCl,
25 25
392.55, mp 183–184 ◦ C, [α]D +25.5◦ (c = M r 375.93, mp 163–168.5 ◦ C, [α]D +59.8◦ (c
25 ◦
5, benzene), [α]D +16 (c = 5, ethanol), d- = 0.6, water).
tartrate [2922-44-3], C25 H32 N2 O2 · C4 H6 O6 , Synthesis: 4-Dimethylamino-3-methyl-1,2-
25
M r 542.64, mp 189–192 ◦ C, [α]D +25.5◦ (c = diphenyl-butan-2-ol is formed by Grignard
25 ◦ reaction of 3-dimethylamino-2-methyl-1-
5, water), [α]D +30.5 (c = 5, CH3 OH).
Synthesis [48, 524]: phenylpropan-1-one with benzylmagnesium
chloride. The preferred product is the α-
diastereomer (75 % α-form = 1SR,2RS isomer,
15 % β-form). The α-form crystallizes and the
diastereomeric β-form remains in solution, be-
cause of its better solubility. Racemic resolution
to obtain the analgetically (+)-enantiomer can
be performed on the pure α-Grignard product
via fractional crystallization of the salts with
d-camphorsulfonic acid. Alternatively the reso-
lution can be achieved by treating the racemic
Mannich product 3-dimethylamino-2-methyl-1-
phenylpropan-1-one with (−)-dibenzoyltartaric
acid in acetone as solvent [582, 585–587] .

Opioid receptor binding: Dextromoramide is


a µ-selective opioid with a higher receptor affin-
ity than morphine.
Clinical use: Dextromoramide tartrate [525]
is a strong opioid, structurally related to
methadone and is used in the treatment of severe
pain. Dextromoramide has a high oral availabil-
ity and is administered orally or rectally. The par-
enteral potency is in the range of morphine, but
the duration of action is shorter. The compound
has a morphine-type abuse and dependence po-
tential.
Trade name: Palfium (France, UK, Nether-
lands).
Analgesics and Antipyretics 63

Opioid receptor binding: Dextropropoxy- Alternatively, diphenylacetonitrile can be


phene has a µ-opioid receptor binding affinity condensed with ethyl 1-(2-chloro-ethyl)-4-
lower than morphine. Binding at other opioid phenylpiperidine-4-carboxylate in the presence
receptors is even weaker. of sodium amide.
Clinical use: Dextropropoxyphene is a mod-
erately potent opioid analgesic often combined
with paracetamol or acetylsalicylic acid. As hy-
drochloride or napsylate it is used orally for the
treatment of mild to moderate pain [529]. Ad-
verse reactions in the therapeutic range are mild
and include drowsiness, dizziness, sedation, and
nausea. Overdosage can induce serious adverse
events including profound sedation, respiratory Opioid receptor binding: Diphenoxylate is
depression, cardiovascular disturbances, con- the active metabolite of difenoxine [532] and
vulsions, and psychotic reactions, often with fa- has a high affinity and selectivity for the µ-type
tal outcome [530]. Dextropropoxyphene has a of opioid receptor.
relatively low abuse liability. Abuse by injection Clinical use: Diphenoxylate [533] is a syn-
is impeded by severe irritations at the injection thetic pethidine derivative with a limited ac-
side. cess to the brain and marginal analgesic activity.
Because of severe and often fatal intoxica- After oral administration, it reduces intestinal
tions the compound is more and more substituted motility and secretion and is used for the treat-
by other weak opioids. ment of diarrhea. Side effects include dizziness,
Trade names: Antalvic (France), Darvon drowsiness, euphoria, anorexia, intestinal paral-
(USA), Develin (Germany), Doloxene (UK). ysis. Prolonged use of high dosages may induce
morphine-like physical dependence. To avoid
Diphenoxylate [915-30-0], ethyl 1- abuse, diphenoxylate is routinely combined with
(3-cyano-3,3-diphenylpropyl)-4-phenylpiperi- small amounts of atropine.
dine-4-carboxylate, C30 H32 N2 O2 , M r 452.59; The compound is now substituted by lop-
hydrochloride [3810-80-8], C30 H32 N2 O2 · eramide, which has less CNS side effects.
HCl, M r 489.06, mp 220.5–222.0 ◦ C. Trade names: Reasec (Germany, Italy),
Synthesis: The reaction of ethyl 4- Diaserd (France), Tropergen (UK), Lomotil
phenylpiperidine-4-carboxylate with 4-bromo- (USA).
2,2-diphenylbutyronitrile yields diphenoxylate
[48, 531]. Loperamide [53179-11-6], 4-[4-(4-chloro-
phenyl)-4-hydroxypiperidin-1-yl]-N,N-dime-
thyl-2,2-diphenylbutyramide, 4-(4-chlorophe-
nyl)-4-hydroxy-N,N-dimethyl-α, α-diphenyl-
1-piperidinebutanamide, C29 H33 CIN2 O2 , M r
477.04; monohydrochloride [34552-83-5],
C29 H33 ClN2 O2 · HCl, M r 513.51, mp 222–
223 ◦ C (decomp.).
Synthesis [48, 534, 535] (see next page).
Opioid receptor binding: Loperamide has a
morphine-like affinity and selectivity for the µ-
opioid receptor [536].
Clinical use: Loperamide [537] is a syn-
thetic opioid subjected to extensive first-pass
metabolization after oral administration. There-
fore little intact drug reaches the systemic cir-
culation and the central nervous system. Lop-
eramide has no centrally mediated analgesic ef-
ficacy, but may have a clinically useful analgesic
64 Analgesics and Antipyretics

effect via peripheral opioid receptors [539]. Sys- Piritramide [302-41-0], 1 -(3-cyano-3,3-
temic and central opioid side effects are widely diphenylpropyl)-[1,4 ]bipiperidinyl-4 -carbox-
missing. Orally administered loperamide acts lo- amide, C27 H34 N4 O, M r 430.59, mp 149–
cally in the gut by inhibition of intestinal motil- 150 ◦ C.
ity and secretion [540]. Besides the strong µ- Synthesis: Piritramide is prepared by conden-
opioid action, calcium and calmoduline antag- sation of 4-piperidinopiperidine-4-carboxamide
onism are involved in the antidiarrheal activ- with 3,3-diphenyl-3-cyanopropylbromide [48,
ity. The compound is used in doses of 4–8 mg 543].
for the treatment of acute and chronic diarrhea
and for the management of colostomies and
ileostomies [538]. Adverse effects include nau-
sea, dry mouth, dizziness. High doses can induce
toxic megacolon and paralytic ileus. The com-
pound has no abuse and dependence potential
and is meanwhile available as over the counter
(OTC) product [542].

Opioid receptor binding: Piritramide is a µ-


opioid with morphine-like affinity and receptor
selectivity.
Clinical use: Piritramide [544] is a synthetic
opioid analgesic used for the treatment of acute,
preferentially postoperative pain. It is given in
doses of 15 mg by the intramuscular, subcuta-
neous, or intravenous route. Piritramide has a
morphine-like effect – side effect profile.
Trade names: Dipidolor (Germany, Belgium,
Netherlands), Piridolan (Sweden).

3.1.2. Other Structures

Tilidine [51931-66-9], ethyl trans-


2-dimethylamino-1-phenylcyclohex-3-ene-
Trade names: Imodium (Germany, Belgium, carboxylate, C17 H23 NO2 , M r 273.17, mp
USA, France), Arret (UK). 34 ◦ C, bp (133.32 Pa) 95.5–96 ◦ C; hydro-
Loperamide is also marketed in many generic chloride [27107-79-5], C17 H23 NO2 · HCl, M r
formulations.
Analgesics and Antipyretics 65

309.84, mp 159 ◦ C; hydrochloride hemihydrate 75 mg for the treatment of moderate to severe


C17 H23 NO2 · HCl · 1/2 H2 O, M r 318.80, mp pain. Tilidine has a substantial abuse potential.
125 ◦ C. To avoid parenteral misuse, oral preparations
Synthesis: Reaction of buta-1,3-dienyl-di- are combined in some countries with a small
methylamine 4 with ethyl atropate 5 yields ti- amount of naloxone hydrochloride. This is in-
lidine as a cis/trans mixture (trans : cis = 2 : 3). tended to induce withdrawal symptoms after il-
Most of the analgetically inactive cis isomer is licit parenteral administration. After oral admin-
separated as a zinc complex and the trans isomer istration, the naloxone component is metabol-
is isolated as the hydrochloride. The cis isomer ically inactivated and does not inhibit analge-
can be epimerized to the trans form by treating sia. After parenteral administration the naloxone
the epimeric mixture with acid [48, 545–548]. component is active and blocks the tilidine effect
and can induce withdrawal reactions [551]. The
combination with naloxone, in contrast to pure
tildine, is available under normal prescription.
Trade names: Valoron (Germany, Switzer-
land), Tilidate (Spain).
Combination with naloxone: Tilidin-N, Val-
oron N.

Tramadol [27203-92-5], cis-2-dimethyl-


aminomethyl-1-(3-methoxyphenyl)cyclohex-
anol, C16 H25 NO2 , M r 263.38; hydrochloride
[36282-47-0], C16 H25 NO2 · HCl, M r 299.84,
mp 180–181 ◦ C.

Opioid Receptor Binding: Tilidine itself has


a low opioid receptor binding affinity, whereas
the active metabolites nortilidine and bisnortili- Synthesis: Grignard reaction of 2-(dimethyl-
dine have a high affinity and selectivity for the aminomethyl)cyclohexanone (obtained by Man-
µ-type of opioid receptor [549]. nich reaction of cyclohexanone, formaldehyde,
Clinical use: Tilidine [550] is rapidly me- and dimethylamine hydrochloride) and the Grig-
tabolized to the active metabolites nortilidine nard reagent of 3-bromoanisole yields tramadol
and bisnortilidine. It can be given orally and as a cis/trans mixture (cis : trans = 85 : 15). Tra-
parenterally and is used in single doses of 50– madol (cis isomer) is separated from the reaction
mixture by crystallization of the hydrochloride
66 Analgesics and Antipyretics

salt. A crystallization process of the hydrate or 3.1.2.1. Mixed Opioid Agonist – Antagonists
hydrobromide salts is also described. The trans and Partial Agonists
isomer can be epimerized to the cis isomer by
strong acids [48, 552, 553]. Mixed agonist – antagonists are opioid com-
Opioid receptor affinity: Tramadol [554] it- pounds that have in vivo relevant affinity for
self has a weak opioid receptor affinity, the ac- more than one type of opioid receptors, com-
tive metabolite O-demethyltramadol shows µ- monly for the µ- and κ-types. The compounds
selective binding with an affinity about ten times of this group are historically derived from the an-
lower than morphine. tagonist nalorphine. At the µ-receptor they act as
Clinical use: Tramadol hydrochloride [555] antagonists or partial agonists with low intrinsic
is a centrally acting analgesic with a µ-opioid activity. At the κ-receptor they act mostly as ag-
and nonopioid action component [556]. The onists. These substances are used as analgesics
nonopioid component acts spinally via inhibi- and the κ-agonistic effect and/or the partial µ-
tion of noradrenaline (NA) and serotonin (5HT) agonistic effect is responsible for their analgesic
reuptake. Tramadol is a racemic mixture of two efficacy. The side effect profile and the misuse
optical enantiomers and uptake inhibition and potential depend on the relative prevalence of
opioid properties are differentially distributed either component in the individual compound.
between the enantiomers [557]. Opioid recep- The κ-component effects dysphoria, hallucina-
tor affinity is low, but like codeine, tramadol tions, and other psychotic reactions, but induces
is biotransformed to the O-demethyl derivative a low level of respiratory depression and ad-
(M1), which has an essentially higher µ-opioid diction. Compounds with a marked κ-agonistic
receptor affinity and may be responsible for the component such as pentazocine are meanwhile
opioid properties of the compound. Tramadol withdrawn from the market because of their un-
is marketed worldwide and has become one of pleasant side effect profile. The µ-component is
the most important centrally acting analgesics responsible for respiratory depression, eupho-
[558]. It is used in single doses of 50–100 mg ria, and dependence potential. Compounds with
for the treatment of acute and chronic medium a more µ-antagonistic profile have a low abuse
to severe pain. The compound has a high oral potential and are not under narcotic drug control,
bioavailability and can be given by mouth, rec- compounds with a stronger µ-agonistic com-
tally, or by intramuscular, subcutaneous, or slow ponent are scheduled. Application of a mixed
intravenous injection or infusion. Typical side agonist-antagonist in an opioid pretreated pa-
effects are nausea, sweating, and dizziness. High tient may reduce analgesia and induce a with-
doses may have a pro-convulsive effect. Tra- drawal reaction.
madol shows a reduced level of opioid side ef-
fects like respiratory depression and constipa- Buprenorphine [52485-79-7], 17-cy-
tion. It has a very limited abuse potential and in clopropylmethyl-α-(1,1-dimethylethyl)-4,5α-
most countries is not subject to narcotic control. epoxy-18,19-dihydro-3-hydroxy-6-methoxy-
Tramadol is used as a mono-compound or as α-methyl-6,14-ethanomorphinan-7-methanol,
a combination with paracetamol [559]. C29 H41 NO4 , M r 467.30, mp 209 ◦ C; hydro-
Trade names: Adolonta (Spain), Contra- chloride [53152-21-9], C29 H41 NO4 · HCl, M r
mal (Belgium, France, Italy), Nobligan (Skan- 504.10.
dinavia), Tramal, Tramal Long (Germany, Synthesis [48, 424–427] (see next page).
Switzerland, Austria), Tramacet (UK), Ultram Opioid receptor binding: Buprenorphine
(USA), Ultracet (USA), Xprim (France), Zydol [560] has a mixed agonistic – antagonistic ac-
(UK, Ireland). tion profile with a high affinity at the µ-, κ-, and
Combination with paracetamol: Ultracet δ-opioid receptor. A ca. 100-fold lower affinity
(USA), Zaldiar (Germany, Austria, Switzer- was observed at the ORL1-receptor. The com-
land). pound shows a slow receptor dissociation which
Tramadol is also marketed in many generic can explain some peculiarities in the pharmaco-
formulations. logical actions [561].
Clinical use: Buprenorphine [562] is used
against moderate to severe pain, mostly for
Analgesics and Antipyretics 67

chronic pain treatment. The potency is about 20– as a matrix patch formulations with release rates
30 times higher than that of morphine. Buprenor- of 35, 52.5, and 70 µg/h.
phine may be used for anesthesia premedication The use of buprenorphine for opioid addic-
or as adjunct to anesthetics. The compound has tion becomes more and more important. For this
a long duration and a slow offset of action and indication, buprenorphine is used in doses up to
is therefore suited for the substitution treatment 24 mg sublingually alone or in combination with
of opioid addiction [563, 564]. Due to its par- naloxone.
tial agonistic properties buprenorphine can act in Adverse reactions of buprenorphine are typi-
combination with full agonists as an antagonist, cal of µ-opioids and include respiratory depres-
reducing their effect and precipitating a with- sion, drowsiness, nausea, and vomiting. Respi-
drawal reaction in opioid dependent persons. ratory depression often occurs delayed, is long
Antagonistic properties are seen in doses much lasting, and may need higher doses of naloxone
higher than the analgesic dose range. There- [566, 567]. Buprenorphine has a limited abuse
fore no special precaution is necessary when potential and withdrawal reactions, due to slow
changing the treatment with a standard opioid receptor dissociation, are mild and delayed.
agonist to buprenorphine or vice versa [565]. Trade names: Buprenex (USA), Buprex,
Buprenorphine is given parenterally, orally (sub- Subutex (Germany, France, USA), Temgesic
lingual), or by the transcutaneous route as a (Germany, France, UK), Transtec (Germany,
patch. The doses for slow intravenous or in- Austria, Switzerland.
tramuscular administration are 300–600 µg, the Combination with naloxone: Suboxone
sublingual doses are 200–400 µg, both given ev- (USA).
ery 6–8 h. Buprenorphine patches are available
68 Analgesics and Antipyretics

Butorphanol [42408-82-2], 11-cyclobutyl- Clinical use: Butorphanol [572, 573] is a


methyl-1,2,3,4,9,10-hexahydro-4α,10-pro- fairly potent opioid analgesic. It is used for the
panophenanthrene-6,10α-diol, C21 H29 NO2 , treatment of moderate to severe pain, for mi-
M r 327.22, mp 215-217 ◦ C, [α]D − 70◦ graine and headache [574] and as an adjunct
(c = 0.1, CH3 OH); tartrate [58786-99-5], to anesthesia. Butorphanol is orally inactive but
C21 H29 NO2 · C4 H6 O6 , M r 477.55, mp 217– can be given by the nasal route [574]. The usual
219 ◦ C, [α]D − 64.0◦ (c = 0.4, CH3 OH). administration is via the intramuscular or intra-
Synthesis [48, 568–570] (see on top). venous route. The intramuscular doses are 1–4
Opioid receptor binding: Butorphanol [571] mg every 3–4 h, the intravenous doses are 0.5–2
is a mixed agonist – antagonist opioid with high mg. Nasal doses are ≈ 1 mg per spray in each
µ- and κ-receptor affinity and full agonistic ac- nostril.
tivity at the κ-receptor and partial agonistic – The plasma elimination half-life of butor-
antagonistic effect at the µ-receptor. phanol is in the range of 3 h. Butorphanol [571]
Analgesics and Antipyretics 69

has a side effect profile combining morphine- ment of moderate to severe pain. It is given by
and pentazocine-like symptoms. They include injection in the dose range of 5–20 mg. Side ef-
drowsiness, weakness, sweating, feelings of fects include nausea, vomiting, and drowsiness.
floating, and nausea. It has respiratory depres- Overdosing can be treated with naloxone. In opi-
sant properties similar to morphine but with a oid pretreated patients, dezocine may precipitate
ceiling effect. As antidote naloxone can be used. withdrawal symptoms.
Overt hallucinations or other psychotic effects Trade name: Dalgan (USA, no longer mar-
are rare and less often reported than with penta- keted).
zocine. The compound has a low abuse potential
and has not been submitted to narcotic control. Meptazinol [54340-58-8], 3-(3-ethyl-
Trade name: Stadol (USA). 1-methylazepan-3-yl)phenol, 3-(3-ethyl-
hexahydro-1-methyl-1H-azepin-3-yl)phenol,
Dezocine [53648-55-8], 15-amino-1- C15 H23 NO, M r 233.35, mp 127.5–133 ◦ C; hy-
methyltricyclo[7.5.1.0127,255 ]pentadeca-2,4,6- drochloride [59263-76-2], C15 H23 NO · HCl,
trien-4-ol, [5R-(5α,11α,13S*)]-13-amino- M r 269.81.
5,6,8,9,10,11,12-octahydro-5-methyl-5,11- Synthesis [48, 581, 582]:
methanobenzocyclodecen-3-ol, C16 H23 NO,
M r 245.36; hydrobromide [57236-36-9],
C16 H23 NO · HBr, M r 326.27, mp 269–270 ◦ C.
Synthesis [48, 575, 576]:

Opioid receptor binding: Dezocine [577] is


a mixed agonist – antagonist with binding affin-
ity to the µ-opioid receptor in the range of mor- Opioid receptor binding: Meptazinol [583] is
phine. The δ- and κ-affinity is 10–100-fold lower a partial µ-agonist with a high µ-receptor affin-
[578]. ity. A selective action at a special subtype of
Clinical use: Dezocine [579, 580] is a µ-opioid receptor (µ-1 subtype, [584]) is con-
medium-potent opioid and is suited for the treat- troversial.
70 Analgesics and Antipyretics

Clinical use: Meptazinol [585] is used par- Synthesis: Starting material for the nal-
enterally (50–100 mg) or orally to treat moder- buphine synthesis is oxycodone (see page 55).
ate or moderately severe pain. The compound After ether cleavage to oxymorphone the prod-
has a shorter duration of action than morphine. uct is acylated and the N-methyl group is re-
The most common side effects are nausea, vom- moved by treatment with cyanogen bromide.
iting, and dizziness. Sweating and hypotension The acetyl groups are hydrolyzed with dilute
can also occur. Meptazinol has opioid antago- hydrochloric acid. The resulting 14-hydroxydi-
nistic properties and can induce withdrawal in hydronormorphinone 6 is N-alkylated with cy-
opioid-dependent persons. The compound has a clobutylmethyl bromide, and the carbonyl group
very low dependence potential and is not under at C-6 is reduced [48, 586, 587].
narcotic drug control. The compound is reported Opioid receptor binding: Nalbuphine is a
to be relatively free of respiratory depressant ac- mixed agonist – antagonist opioid with a high
tivity, which was attributed to selective binding affinity at the µ- and κ-opioid receptor. At the
to a µ-1 subtype of the opioid receptor [584]. κ-receptor the compound is an agonist, at the µ-
This is not confirmed by others and the effect receptor a partial agonist with a very low intrin-
may be alternatively explained by a choliner- sic activity, thus acting more as a µ-antagonist
gic action component [583] of the compound. [588].
In accordance with a low κ-receptor affinity, the Clinical use: Nalbuphine [589, 590] is an opi-
incidence of psychotomimetic actions and hal- oid analgesic with a mixed agonist – antagonist
lucinations is low. action profile. Analgesia is limited by a “ceiling”
Trade name: Meptid (Germany, UK). effect. The compound has a low oral availability
and is used as hydrochloride only for parenteral
Nalbuphine [20594-83-6], (5α6α)-17-(cy- application to treat moderate to severe pain and
clobutylmethyl)-4,5-epoxymorphinan-3,6,14- as adjunct to anesthesia. The dose for pain re-
triol, C21 H27 NO4 , M r 357.44, mp 230.5 ◦ C; hy- lief is 10–20 mg. The most frequent side effects
drochloride [23277-43-2], C21 H27 NO4 · HCl, are drowsiness, sweating, nausea, and vomiting.
M r 393.91, mp 291–292 ◦ C (decomp.). Hallucinations and psychotomimetic reactions
are less frequent than with pentazocine, reflect-
ing a relative weak κ-agonistic component of the
compound. Nalbuphine induced respiratory de-
pression is less severe than with morphine and is
limited by a ceiling effect [591]. The dependence
potential is low and nalbuphine is not subject to
narcotic control.
Trade name: Nubain (Germany, France, UK,
USA).

Nalorphine [62-67-9], (5α,6α)-7,8-dide-


hydro-4,5-epoxy-17-(2-propenyl)morphinan-
3,6-diol, C19 H21 NO3 , M r 311.38, mp 208–
25
209 ◦ C, [α]D −155.3◦ (c = 3, CH3 OH); hy-
drochloride [57-29-4], C19 H21 NO3 · HCl, M r
347.84, mp 260–263 ◦ C; hydrobromide [1041-
90-3], C19 H21 NO3 · HBr, M r 392.29, mp 258–
259 ◦ C (decomp.).
Synthesis: Diacetylmorphine (heroin) is
demethylated with cyanogen bromide and hy-
drolyzed to normorphine, which is alkylated
with allyl bromide to give nalorphine [48, 592].
Analgesics and Antipyretics 71

enteral, and rectal formulations for the treatment


of moderate to severe pain. Pentazocine induces
morphine-type side effects like dizziness, nau-
sea, vomiting, sedation, and sweating. In ad-
dition, it can cause κ-agonist type psychoto-
mimetic effects like hallucinations, nightmares
and thought disturbances. Respiratory depres-
sion is weaker than with morphine and is subject
to a “ceiling” effect.
The compound has a relevant abuse potential
[597] and high doses may produce dependence
of the morphine type. Pentazocine effects can be
antagonized by naloxone.
Because of its side effect profile, the com-
pound is no longer used.

Opioid receptor binding: Nalorphine [113] is


a mixed agonist – antagonist with a high affinity
and low intrinsic action at the µ- and κ-opioid
receptors.
Clinical use: Despite a predominant antago-
nistic action profile nalorphine has substantial
analgesic activity and was the first opioid com-
bining analgesic activity with an antagonistic ac-
tion. The compound is no longer used because of
κ-agonistic type adverse reactions such as anx-
iety, hallucinations, and dysphoric mood alter-
ations.

Pentazocine [359-83-1], 6,11-di-


methyl-3-(3-methylbut-2-enyl)-1,2,3,4,5,6-
hexahydro-2,6-methanobenzo[d]azocin-8-ol,
(2α,6α,11R*)-1,2,3,4,5,6-hexahydro-6,11-di-
methyl-3-(3-methyl-2-butenyl)-2,6-methano-
3-benzazocin-8-ol, C19 H27 NO, M r 285.42,
mp 145–148 ◦ C; hydrochloride [2276-52-
0], C19 H27 NO · HCl, M r 321.89; lactate (1 :
1) [17146-95-1], C19 H27 NO · C3 H6 O3 , M r
375.51.
Synthesis [48, 594, 595] (see right column).
Opioid receptor binding: Pentazocine [596]
is a mixed opioid agonist – antagonist with ag-
onistic effects at the κ- and partial antagonistic
effects at the µ-type of opioid receptor.
Clinical use: Pentazocine is a potent anal-
gesic with therapeutic dosages in the range of
25–100 mg. It is orally bioavailable and was
used as hydrochloride or lactate in oral, par-
72 Analgesics and Antipyretics

3.1.2.2. Opioid Antagonists [598]

Opioid and narcotic antagonists (Table 2) are


compounds with binding affinity at opioid re-
ceptors, but without intrinsic activity. They only
block the receptor without inducing a genuine
effect but are able to inhibit or reverse the ac-
tions of opioid agonists or partial agonists. An-
tagonists may or may not be selective for one
opioid receptor type. Only antagonists with a
high affinity and/or selectivity for the µ-receptor
have clinical application. They are used to termi-
nate medically desired effects of µ-opioid such
as anesthesia, to reverse side effects like respira-
tory depression or to treat opioid intoxications.
Acute administration of antagonists precipitates
withdrawal reactions in opioid-dependent sub-
jects. In the course of long-term treatment orally
active antagonists are used for detoxification of
opioid addicts.
Antagonists with δ- or κ-receptor selectivity
have no clinical application but are widely used
in opioid research.

Levallorphan [152-02-3], 17-(2-propenyl)- Naloxone [465-65-6], (5α)-4,5-epoxy-


morphinan-3-ol, 1-N-allyl-3-hydroxymorphi- 3,14-dihydroxy-17-(2-propenyl)morphinan-6-
nane, C19 H25 NO, M r 283.42, mp 180–182 ◦ C, one, C19 H21 NO4 , M r 327.37, mp 184 ◦ C (also
25
[α]D −88.9 ◦ (CH3 OH); hydrogen tartrate (1 : 20
reported as 177–178 ◦ C), [α]D −194.5◦ (c
1) [71-82-9], C19 H25 NO · C4 H6 O6 , M r 433.50, = 0.93, CHCl3 ); hydrochloride [357-08-4],
25
mp 176–177 ◦ C, [α]D −39◦ (water). C19 H21 NO4 · HCl, M r 363.84, mp 200–205 ◦ C.
Synthesis: Starting material for the lev- Synthesis: 14-Hydroxydihydronormorphinone
allorphan synthesis is 1-(4-methoxybenzyl)- 6 (see page 70) is N-alkylated with allyl bro-
1,2,3,4,5,6,7,8-octahydro isoquinoline (1) mide [48, 606, 607].
(preparation see page 54) [48, 599–602].
Opioid receptor binding: Levallorphan has a
high binding affinity but a low intrinsic activity
at the µ-opioid receptor.
Analgesic efficacy and clinical use: Levallor-
phan [603] is an opioid antagonist with practi-
cally no analgesic action. It has been used as
one of the first relative pure antagonists for the
treatment of opioid overdosage, to reverse opi-
oid central depression and to antagonize opioid- Opioid receptor binding: Naloxone [608] is
induced respiratory impairment [604]. The com- a pure opioid antagonist with a high affinity and
pound is now replaced by naloxone [605]. a limited selectivity for the µ-receptor.
Analgesics and Antipyretics 73

Efficacy and clinical use: Naloxone [609, b)


610] has no analgesic activity. The compound
is the standard antidote to treat opioid adverse
reactions, opioid overdoses, or to stop an in-
tended use of an opioid compound. Typical in-
dications are inhibition of opioid-induced res-
piratory depression, termination of opioid anes-
thesia or protection of neonates following opi-
oid treatment during labor. Naloxone has a short
duration of action and repetitive administration
may be necessary to antagonize longer acting
agonists. To avoid parenteral misuse of non-
scheduled oral opioid formulations (tilidine,
pentazcocine), a small amount of naloxone is
added which is orally inactivated, but is fully
active after parenteral administration.
Naloxone is orally inactive and is only used
parenterally in single or repetitive doses of 0.4–
2 mg up to a total dose of 10 mg, as an intra- Opioid receptor binding: Naltrexone has a
venous bolus injection or by infusion. The com- high affinity and selectivity for the µ-opioid re-
pound is more potent against pure opioid ag- ceptor [612].
onists than against mixed agonist – antagonists. Efficacy and clinical use: Naltrexone [613,
Caution should be used in opioid-dependent per- 614] is a pure opioid antagonist and has no
sons or in persons under high-dose opioid treat- analgesic activity. Naltrexone has a higher in-
ment, as naloxone may precipitate an acute with- travenous potency and longer duration of action
drawal reaction. Naloxone is relatively free of than naloxone. It has a higher oral bioavailability
side effects. Nausea, vomiting, and convulsions and is given by mouth to treat opioid dependence
have occasionally been reported. and to maintain abstinence during opioid detoxi-
Trade names: Narcan (France, UK, USA), fication [615]. In opioid-dependent persons, nal-
Narcanti (Germany, Austria). trexone induces an acute withdrawal reaction.
Trade names: Nalorex (France, UK), Ne-
Naltrexone [16590-41-3], (5α)-17-(cy- mexin (Germany), Revia (USA), Trexan (USA).
clopropylmethyl)-4,5-epoxy-3,14-dihydroxy
morphinan-6-one, C20 H23 NO4 , M r 341.40, N-Methylnaltrexone bromide [073232-
mp 168–170 ◦ C; hydrochloride [16676-29-2], 52-7], 17-(cyclopropylmethyl)-4,5a-epoxy-
C20 H23 NO4 · HCl, M r 377.87, mp 274–276 ◦ C. 3,14-dihydroxy-17-methyl-6-oxomorphi-
Synthesis: nanium bromide, C21 H26 BrNO4 , M r 436.34,
mp 251 ◦ C, [α]D25 - ?◦ (c = 1.49, CH3 OH);
a) Naltrexone is prepared in the same way as N-methylnaltrexone iodide [073232-53-8],
naloxone starting from oxycodone via the in- C21 H26 INO4 , M r 483.34.
termediate 6 except that the nitrogen atom Synthesis: Methylnaltrexone is obtained
is alkylated with cyclopropyl bromide [48, by quaternization of N-(cyclopropylmethyl)-
611]. noroxymorphone (naltrexone) with methyl bro-
mide in acetone/dimethylformamide [616]:
74 Analgesics and Antipyretics

Efficacy and intended clinical use: N- neuropathic pain as pain initiated or caused by
Methylnaltrexone [617] is the polar derivative a primary lesion or dysfunction in the nervous
of naltrexone, which does not reach the central system. This malfunction of the nervous system
nervous system but can block intestinal opioid can occur at any point along the sensory path-
receptors. The compound is under development way (peripheral nerve, dorsal root ganglion, and
to counteract opioid induced bowel dysfunction CNS). The term peripheral neuropathic pain is
such as constipation and megacolon [618]. usually used to refer to abnormalities in the pe-
ripheral nervous system. Central pain is retained
Nalmefene [055096-26-9], N-(cyclopro- as the term when the lesion or dysfunction oc-
pylmethyl)-4,5α-6-methylenemorphinan-3,1β- curs in CNS.
diol, C21 H25 NO3 , M r 339.43, mp 105–110 ◦ C;
hydrochloride [58895-64-0], C21 H26 ClNO3 , Table 4. Etiological classification of neuropathic pain [623]
M r 375.89, mp 188–190 ◦ C. Injury Examples
Synthesis: Nalmefene is synthesized by a Infection/inflammation
Wittig reaction of naltrexone with triphenyl- • postherpetic neuralgia
methylphosphonium bromide in DMSO under • HIV

basic catalysis of NaH. Trauma


• post-amputation phantom limb
pain
• spinal cord injury
• surgery

Ischemia
• diabetic neuropathy
• central post-stroke pain

Compression
Nalmefene is a 6-methylene analogue of nal- • sciatica
• carpal tunnel
trexone. It is a pure opioid receptor antagonist • trigeminal neuralgia
and has a high affinity for the κ-opioid receptor.
Nalmefene has a long duration of action and is Neoplasm
used in oral and parenteral formulations. • invasion of neural structures
Nalmefene is used to reverse opioid intoxica- • compression by tumor
expansion
tion [619] and for the treatment of opioid depen-
dence [620]. The compound is in clinical devel- Iatrogenic
opment for the treatment of alcoholism [621], to • vinca alkaloids
support smoking cessation and to control patho- • antiretrovirals
• post-irradiation
logical gambling [622].
Trade name: Revex (USA). Idiopathic
• multiple sclerosis
• trigeminal neuralgia
• complex regional pain
3.2. Antineuropathic Analgesics and syndrome
Other Non-opioid Compounds
Classification: Neuropathic pain has been
3.2.1. Neuropathic Pain Treatment
traditionally classified according to its etiology
[623–629, 658, 659]
(Table 1) and tends to be chronic and unremit-
ting. Patients with neuropathic pain are often dif-
Definition: Neuropathic pain results from dam-
ficult to treat, the pain is sometimes relatively un-
age to the nervous system due to many diverse
responsive to conventional analgesics and thus
processes. The damage causes persistent, dis-
a wide variety of drugs from many classes have
tressing pain, that is reputedly unresponsive to
been suggested for this indication. Table 4. indi-
conventional analgesics. The International As-
cates that neuropathic pain refers to a group of
sociation for the Study of Pain (IASP) defines
Analgesics and Antipyretics 75

painful disorders characterized by pain due to fer the best treatment options. Other interven-
dysfunction or disease of the nervous system at tions such as stimulation, blockade, or destruc-
a peripheral level, a central level, or both. In con- tion of nerves and psychological treatments are
trast to nociceptive pain, which results from the also part of the armament for treating neuro-
activation of pain receptors (nociceptors), neu- pathic pain. Table 5. gives an overview on the
ropathic pain is the result of injury to the pain- current available evidence-based treatment op-
conducting nervous system. tions for neuropathic pain.
Symptoms: Neuropathic pain is a complex en-
tity with many symptoms and signs that fluctu- Table 5. Evidence-based treatment options for neuropathic pain
ate, in number and intensity, with time. The de- [623]

scriptors associated with neuropathic pain have Drug class Proposed mechanism Examples
of action
been described qualitatively as burning, stab- Tricyclic
bing, shooting, aching and electric shock-like, antidepressants
• unspecific sodium • amitryptiline
amongst others. Pain may be continuous or channel blockade • nortriptyline
paroxysmal and felt superficially or deeply. It • inhibition of • desipramine
norepinephrine • imipramine
may also occur spontaneously, independent of a and serotonin
stimulus. Sensory changes in the injured area reuptake
are common. There may be areas of sensory • blockade of
α-adrenergic
loss but also of altered sensation to cutaneous receptors
stimuli. Non-noxious stimuli, e.g. stroking, may
elicit pain (allodynia). Mildly painful stimuli Serotonin and/or blockade of serotonin
norepinephrine and/or norepinephrine
may cause excessive pain (hyperalgesia) and reuptake inhibitors reuptake • duloxetine
• venlafaxine
repetitive stimulation can cause an explosive
build-up of pain that lasts well beyond the stim- Anticonvulsants
ulus (hyperpathia). These sensory abnormalities • unselective block • benzodiazepines
can be classified by type of stimulus – thermal, of sodium • carbamazepine
channels • oxacarbazepine
mechanical, and chemical. Mechanically evoked • phenytoin
events can be further subdivided into dynamic • lamotrigin
(brush-stroke evoked), static (pressure-evoked) • topiramate
• zonisamide
and punctate.
Anticonvulsants
Current Treatment Options in Neuro- • selective block of • gabapentin
pathic Pain. There are several rationales to calcium channels • pregabalin

guide treatment of patients with neuropathic Local anesthetics


pain. The most widely used at present is based on (type 1b
• unspecific sodium • lidocaine
antiarrythmic
the etiology and anatomical distribution of the drugs) (and/or calcium) • mexiletine
pain. Others have suggested a symptom-based channel blockade • bupivacaine
approach on the premise that certain symptoms Drugs for the
might respond best to specific classes of drugs. treatment of
neurological • antagonism of the • memantine
In clinical practise it is shown that one mecha- disorders NMDA receptor • dextromethorphan
nism may lead to different symptoms and that • ketamine
a similar pain symptom or deficit may be pro- Opioids
duced by several different underlying mecha-
• opioid agonism • blockade of
nisms. At present, there is no single treatment norepinephrine
that adequately and predictably controls neuro- reuptake
• increased
pathic pain, nor does there seem to be any way serotonin release
of preventing the development of neuropathic • morphine
pain following nerve injury. Management by a • tramadol
• methadone
combination of both pharmacological and non-
pharmacological approaches, ideally in a mul-
tidisciplinary pain clinic setting, seems to of-
76 Analgesics and Antipyretics

3.2.2. Individual Compounds

Baclofen [1134-47-0], β-(aminomethyl)-


4-chlorobenzenepropanoic acid, δ-(amino-
methyl)-p-chlorohydrocinnamic acid, γ-amino-
β-(p-chlorophenyl)butyric acid, C10 H12 ClNO2 ,
M r 213.66, mp 206-208 ◦ C; hydrochloride,
C10 H13 Cl2 N, M r 250.12, mp 179-181◦ C.
Synthesis:
a.) Baclofen is synthesized by reaction
of 4-chlorobenzaldehyde with ethyl aceton-
acetonate under catalysis of sodium ethoxide.
The resulting condensation product is hydro-
lyzed to 3-(4-chlorophenyl)glutaric acid. Cy-
clization to 3-(4-chlorophenyl)glutaric anhy-
dride and subsequent aminolysis yields 3-
(4-chlorophenyl)glutarimide, which is finally Clinical use: Baclofen, a GABAB agonist,
transformed to baclofen [630, 631] was initially launched in the 1970s for the oral or
parenteral treatment of voluntary muscle spas-
ticity due to cerebrovascular accidents, cere-
bral palsy, meningitis, multiple sclerosis (MS),
spinal lesions, and head injury. Baclofen medi-
ates its effect directly via the GABAB receptor.
An analgesic action of baclofen administered
systemically was first described more than 25
years ago and subsequently confirmed by many
different groups. Despite the large volume of
preclinical data that is now available, the use of
baclofen as an analgesic in humans has been lim-
ited owing, in part, to rapid tolerance and adverse
effects following systemic administration. Pos-
sibly, the only type of pain for which baclofen
can be used systemically is trigeminal neural-
gia. However, when administered intrathecally,
baclofen is able to reduce central pain such as
occurs in stroke [632].
Trade names: Gabalon, Lioresal (Germany,
UK, Italy), Liorésal (France).

Carbamazepine [298-46-4], dibenzo[b,f ]-


azepine-5-carboxamide, C15 H12 N2 O, M r
236.27, mp 204–206 ◦ C (also reported as 190–
193 ◦ C).
Synthesis [633]:

b.) Alternatively, baclofen can be obtained


by reaction of ethyl-4-chlorocinnamate with
nitromethane followed by hydrogenation and es-
ter cleavage [630, 631].
Analgesics and Antipyretics 77

Alternatively, 5H-dibenzo[b,f ]azepine can Duloxetine [116539-59-4], (+)-(S)-


be treated directly with potassium cyanate yield- N-methyl-N-[3-(naphthalen-1-yloxy)-3-(2-
ing carbamazepine [634]. thienyl)propyl]amine, (S)-N-methyl-γ-(1-
Clinical use: Carbamazepine is an orally ac- naphthalenoxy)-2-thiophenepropanamine
tive anticonvulsant. It is used additionally in the C18 H19 NOS, M r 297.41, mp 118–122 ◦ C,
20
treatment of manic depression and as an anal- [α]D +122 ◦ (c = 1.0, CH3 OH); hydrochloride
gesic in trigeminal neuralgia and other neuro- [136434-34-9], C18 H20 ClNOS5 , M r 333.88,
pathic pain conditions [635]. Common side ef- mp 163–167 ◦ C.
fects are dizziness, drowsiness, ataxia, visual Synthesis: Reaction of 2-acetylthiophene
disturbances, dry mouth, abdominal pain, nau- with paraformaldehyde and dimethylamine in
sea, and vomiting. Other adverse reactions in- ethanol gives 3-(dimethylamino)-1-(2-thienyl)-
clude headache, arrhythmias, and heart block. 1-propanone, which is enantioselectively re-
Trade names: Tegretal (Germany, Italy), duced with a 2:1 complex of (2R,3S)-4-(di-
Tegretol (UK, France, USA). methylamino)-3-methyl-1,2-diphenyl-2-buta-
nol and LiAlH4 in toluene to yield (S)-3-(di-
Clonidine [4205-90-7], (2,6-dichloro- methylamino)-1-(2-thienyl)-1-propanol. The
phenyl)-(4,5-dihydro-1H-imidazol-2-yl)amine, condensation of (S)-3-(dimethylamino)-1-(2-
C9 H9 Cl2 N3 , M r 230.09, mp 139 ◦ C; monohyd- thienyl)-1-propanol with 1-fluoronaphthalene
rochloride [4205-91-8], C9 H9 Cl2 N3 · HCl, M r catalyzed by NaH in DMSO affords the corre-
266.55, mp 305 ◦ C. sponding naphthyl ether (S)-N,N-dimethyl-3-
Synthesis [48, 636, 637]: (naphthalen-1-yloxy)-3-(thiophen-2-yl)propan-
1-amine, which is finally monodemethylated
with 2,2,2-trichloroethyl chloroformate and zinc
in toluene and treated with oxalic acid [641,
642].

Clinical use: Clonidine is an α2 -adrenergic


agonist and is primarily used in the cardiovas-
cular field for blood pressure reduction (→ An-
tihypertensives, Section 5.1.2) [638]. The com-
pound induces analgesia via central α2 -receptor
interaction and can be used orally, parenter-
ally or epidurally for pain treatment, often in
combination with opioids or local anesthetics
[639]. Epidural clonidine – opioid combinations
are preferentially used for neuropathically main-
tained cancer pain. The compound is addition-
ally used for migraine prophylaxis and to reduce
opioid and alcohol withdrawal symptoms [640].
Clonidine frequently induces side effects like
hypotension, sedation, drowsiness, dry mouth,
and constipation. The compound is well ab-
sorbed orally. The ketone intermediate 3-(dimethylami-
Trade names: Duraclon (USA), Catapresan no)-1-(2-thienyl)-1-propanone can also be re-
(Germany), Catapres (USA).
78 Analgesics and Antipyretics

duced with NaBH4 in ethanol to the racemic


alcohol 3-(dimethylamino)-1-(thiophen-2-yl)-
propan-1-ol, which is submitted to optical reso-
lution with (S)-(+)-mandelic acid to provide the
(S)-enantiomer [641, 642].

Reaction of thiophene-2-carboxylic acid with


oxalyl chloride and triphenylphosphine gives
the corresponding acyl chloride thiophene-2-
carbonyl chloride, which is condensed with
vinyltributylstannane, yielding 1-(2-thienyl)-2-
propen-1-one. The addition of HCl to the double
bond of 1-(2-thienyl)-2-propen-1-one affords
3-chloro-1-(2-thienyl)-1-propanone, which is
reduced with BH3 and the chiral (R)-
oxazaborolidine catalyst in THF to give (S)-3-
chloro-1-(2-thienyl)-1-propanol. Treatment of
(S)-3-chloro-1-(2-thienyl)-1-propanol with NaI
in acetone affords the (S)-3-iodopropanol Clinical use: Duloxetine hydrochloride is an
derivative, which is condensed with methy- inhibitor of 5-HT reuptake and norepinephrine
lamine in THF to provide (S)-3-(methylami- reuptake. The compound was launched in 2004
no)-1-(2-thienyl)-1-propanol. Finally, this com- by originator Lilly for the treatment of major de-
pound is condensed with 1-fluoronaphthalene by pression, urinary incontinence, and pain caused
means of NaH in DMA [641, 642]. by diabetic peripheral neuropathy [643–645].
The Friedel – Crafts condensation of thio- The drug is also undergoing phase III clini-
phene with 3-chloropropionyl chloride in pres- cal trials at Lilly for the treatment of generalized
ence of SnCl4 in benzene gives the afore- anxiety and fibromyalgia.
mentioned 3-chloro-1-(2-thienyl)-1-propanone, Trade names: Cymbalta (Eli Lilly, UK,
which is reduced with NaBH4 in ethanol to France, Italy, Germany), Yentrere (Eli Lilly,
yield the racemic alcohol 3-chloro-1-(thiophen- USA, UK, France, Italy, Germany), Arichaim
2-yl)propan-1-ol. Optical resolution of 3- (Boehringer Ingelheim, UK, France, Italy, Ger-
chloro-1-(thiophen-2-yl)propan-1-ol performed many), Xeristar (Boehringer Ingelheim, UK,
by means of immobilized CALB (Novozyme France, Italy, Germany).
435, Novo-Nordisk A/S) affords the enan-
tiomer (S)-3-chloro-1-(thiophen-2-yl)propan-1-
ol [643].
Analgesics and Antipyretics 79

Flupirtine [56995-20-1], ethyl[2-ami- yield the half ester 2-[1-(methoxycarbonyl)cy-


no-6-(4-fluorobenzylamino)pyridin-3-yl]carb- clohexyl]acetic acid, which is subjected to a Cur-
amate, C15 H17 FN4 O2 , M r 304.32, mp tius type rearrangement to give the isocyanate
115–116 ◦ C; monohydrochloride [33400-45- 2-[1-(isocyanatomethyl)cyclohexyl]acetic acid.
2], C15 H17 FN4 O2 · HCl, M r 340.79, mp The desired compound is obtained by hydroly-
214–215 ◦ C; maleate (1 : 1) [75507-68-5], sis of 2-[1-(isocyanatomethyl)cyclohexyl]acetic
C15 H17 FN4 O2 · C4 H4 O4 , M r 420.39, mp acid with HCl, followed by hydrochloric salt re-
175.5–176 ◦ C. moval via anion exchange [651, 652].
Synthesis [48, 646–649]:

Clinical use: Flupirtine maleate [650] is an


analgesic with an unknown mode of action. It is
used in single doses of 100 mg orally and 150
mg rectally for the treatment of different pain The anhydride can also be treated alter-
conditions. natively with N-hydroxylamine to afford the
Trade name: Katadolon (Germany). N-hydroxyimide, which is converted to the
N-benzenesulfonyloxylimide by reaction with
Gabapentin benzenesulfonyl chloride. Subsequent reac-
Synthesis [651, 653–655]: In the original syn- tion of the N-benzenesulfonyloxylimide with
thesis (Goedecke) cyclohexenone is reacted with triethylamine in methanol gives the ure-
ethyl cyanoacetate in the presence of ammo- thane ester methyl 1-(methoxycarbonyl)cyclo-
nia to yield the Guareschi salt, which is hy- hexanecarboxylate, which is submitted to HCl
drolyzed and decarboxylated to give 1,1-cyclo- hydrolysis to afford the hydrochloride of
hexanediacetic acid which is transformed by to gabapentin. The free amino acid is obtained from
the corresponding anhydride with acetic anhy- the hydrochloride via anion exchange.
dride. This anhydride is treated with methanol to
80 Analgesics and Antipyretics

The condensation of cyclohexene-1-


carbaldehyde with benzoylhydrazide in
dichloromethane gives the correspond-
ing hydrazone (E)-N  -(cyclohexenylmethy-
lene)benzohydrazide, which is condensated un-
der reductive conditions with trichloroacetyl
chloride under catalysis of BH3 /(CH3 )2 NH and
Ms-OH (methanesulfonic acid) in diethyl ether
to yield the adduct N  -(cyclohexenylmethyl)-
N  -(2,2,2-trichloroacetyl)benzohydrazide. The
cyclization of this adduct by means of CuCl
and tetramethylenediamine (TMDA) in hot ace-
tonitrile affords the spiro pyrrolidone, which
is dechlorinated and debenzamidated by treat-
ment with wet Raney nickel in ethanol at 110 ◦ C
to provide 2-azaspiro[4,5]decan-3-one. Finally,
this compound is hydrolyzed with hot aqueous
HCl to form the target gabapentin [653–655].
The anhydride can also be treated with am-
monia to yield cyclohexane-1,1-diacetic acid
monoamide. In this route the compound is sub-
mitted to a Hoffman rearrangement to provide
the target amino acid.

The reaction of 2-[1-(carbamoylmethyl)cy-


clohexyl]acetic acid with NaOCl and NaOH
(a Hofmann rearrangement) gives the isocya- Clinical use: Gabapentin was developed as
nate intermediate, which without isolation hy- a structural GABA analogue but it has no di-
drolyzes to the target gabapentin. rect GABAergic action and it does not affect
GABA uptake or metabolism. Preliminary evi-
dence points to the possible effect of gabapentin
on the α2δ type of calcium channels.
The complex pharmacological mechanism
by which gabapentin and the related compound
pregabalin exert their clinical effect in epilepsy
and neuropathic pain, remains unclear [656,
657].
Gabapentin has a clearly demonstrated ef-
ficacy in relieving pain and associated symp-
Analgesics and Antipyretics 81

toms in patients with postdiabetic and posther-


petic neuropathy. The starting dosage which can
be used is 300 mg three times a day. Increase
by 200 mg increments to 200–400 mg three to
four times a day is recommended. As main side
effects, sedation, dizziness, fatique, and pedal
edema are reported [658, 659].
Trade name: Neurontin (Germany).
Gabapentin is also used in many generic for-
mulations.

Lamotrigine [084057-84-1], 6-(2,3-di-


chlorophenyl)-1,2,4-triazine-3,5-diamine, 3,5-
Lamotrigine can also be obtained by the
diamino-6-(2,3-dichlorophenyl)-1,2,4-triazine
following route: Hydrogenation of 2,3-di-
C9 H7 Cl2 N5 , M r 256.09, mp 216-218 ◦ C.
chloronitrobenzene with H2 over Raney nickel
Synthesis: The reaction of the Grignard com-
in methanol gives 2,3-dichloroaniline, which is
pound of 2,3-dichloroiodobenzene with CO2
diazotized with NaNO2 and HCl and treated
in diethyl ether gives 2,3-dichlorobenzoic acid,
with CuCN to yield 2,3-dichlorobenzonitrile.
which is converted to the corresponding acyl
Hydrolysis of the nitrile with NaOH in reflux-
chloride by refluxing with SOCl2 . The re-
ing methanol/water affords 2,3-dichlorobenzoic
action of 2,3-dichlorobenzoyl chloride with
acid, which is treated with hot SOCl2 to pro-
cuprous cyanide and KI in refluxing xylene
vide the corresponding acyl chloride. Reaction
yields 2,3-dichlorobenzoyl cyanide. Finally, this
of 2,3-dichlorobenzoyl chloride with CuCN and
compound is cyclized with aminoguanidine in
KI in refluxing chlorobenzene gives 2,3-dichlo-
DMSO to yield lamotrigine [660–663].
robenzoyl cyanide, which is condensed with
aminoguanidine by means of H2 SO4 /TsOH in
hot toluene to yield 2,3-dichlorobenzoyl cyanide
amidinohydrazone. Finally, this compound is
cyclized by treatment with NaOMe in refluxing
methanol to yield lamotrigine.

Condensation of 2,3-dichlorobenzoyl
cyanide with aminoguanidine in the presence
of polyphosphoric acid (PPA) as catalyst in hot
acetonitrile yields 2,3-dichlorobenzoylcyanide
amidinohydrazone, which is cyclized to the
target 1,2,4-triazine by heating in refluxing
propanol with or without DMSO.
82 Analgesics and Antipyretics

Clinical use: Lamotrigine is a phenyltriazine Alternative route:


derivative and one of the newer antiepilep-
tic agents, that stabilize neural membranes
by blocking the activation of voltage-sensitive
sodium channels and inhibiting the presynaptic
release of glutamate. Lamotrigine was initially
approved in the USA as add-on therapeutic for
partial complex seizures. At doses of 50 to 400
mg/d lamotrigine has demonstrated efficacy in
relieving pain in patients with trigeminal neu-
ralgia refractory to other treatments, such as
carbamazepine, phenytoin, or both. Pain reduc-
tion usually occurs at oral doses of more than 200
mg daily. However, lamotrigine must be dosed
very slowly during the first six weeks of treat-
ment to prevent a toxic rash. Recent negative
results from a clinical trial indicated that more
well-designed trials are needed to better define
the role of lamotrigine as an analgesic drug.
Main side effects are rash, dizziness, unsteadi-
ness, and drowsiness [658, 659, 664].
Trade name: Lamictal (Germany, UK,
France, Italy, USA).

Nefopam [13669-70-0], 5-methyl-1-


phenyl-3,4,5,6-tetrahydro-1H-benzo[f ][1,4]-
oxazocine, C17 H19 NO, M r 253.34; hydro-
chloride [23327-57-3], C17 H19 NO · HCl, M r
289.81, mp 238–242 ◦ C.
Synthesis [48, 665, 666]:

Clinical use: Nefopam [667] is a centrally


acting analgesic without opioid properties. It in-
duces inhibition of NA and 5-HT reuptake and
has antimuscarinic and sympatomimetic actions
which all might be relevant for its analgesic ac-
tion. Nefopam is used orally and parenterally for
the treatment of moderately severe pain. Side
effects are nausea, vomiting, sweating, nervous-
ness, tachycardia, and occasionally convulsions.
Nefopam should not be used in combination
with MAO inhibitors.
Trade names: Ajan (Germany), Acupan (Bel-
gium, France, Italy, UK).

Phenytoin [561-27-2], diphenylhydantoin,


5,5-diphenyl-2,4-imidazolidinedione
C15 H12 N2 O2 , M r 252.27, mp 295-298 ◦ C;
monosodium salt [690-93-3], C15 H11 N2 NaO2 ,
M r 274.26.
Analgesics and Antipyretics 83

Synthesis: Trade names: Epanutin, Phenhydan, Zen-


tropil (Germany), Dihydan (France), Epanutin
a) Phenytoin is synthesized by cyclization of
(UK), Dilantin (USA).
benzil with urea [668, 670].
Topiramate [097240-79-4], 2,3;4,5-bis-O-
(1-methylethylidene)-β-d-fructopyranose sulfa-
mate, 2,3;:4,5-bis-O-(1-methylethylidene)-1-O-
sulfamoyl-β-d-fructopyranose, C12 H21 NO8 S,
25
M r 339.36, mp 122-126 ◦ C, [α]D −34 ◦ (c =
0.4, CH3 OH).
Synthesis: Topiramate is synthesized by two
methods [671, 672]: d-Fructose is reacted with
b) Alternatively phenytoin can also obtained by acetone to produce the thermodynamically more
using benzophenone as a starting material stable bisacetonide. This bisacetonide is then
[669, 670]: condensed with sulfamoyl chloride in the pres-
ence of sodium hydride. Alternatively, the in-
Phenytoin termediate bisacetonide is reacted with sulfuryl
chloride and pyridine in methylene chloride and
then with sodium azide in acetonitrile to furnish
the azido sulfate, which is reduced with copper
powder in methanol [671, 672]

Clinical use: Phenytoin became the first anti-


convulsant to be used to treat neuropathic pain.
The first clinical trials started in 1977. Current
data indicate that the analgesic effect of pheny-
toin is achieved through the blockage of sodium
channels, inhibition of presynaptic glutamate re-
lease and suppression of spontaneous neuronal
ectopic discharges. Phenytoin is also used as
a co-analgesic in combination therapy. As an Alternatively the reaction of diacetone fruc-
oral starting dose, 200 mg at bedtime is recom- tose with sulfamide in presence of of pyridine
mended. An increase by 100 mg increments to in xylene at 128-133 ◦ C yields the target topira-
300–400 mg daily divided in 1–2 doses is pos- mate [673, 674].
sible. Main side effects are slurred speech, rash,
unsteadiness, confusion, or blurry vision [658,
659].
84 Analgesics and Antipyretics

The compound had been under development


for the treatment of neuropathic pain, bipolar
disorders, obesity, pathological gambling, abuse
and dependency, and for smoking cessation.
Trade names: Epitomax, Topamax, Topimax.

Ziconitide [097240-79-4], H-Cys-Lys-


Gly-Lys-Gly-Ala-Lys-Cys-Ser-Arg-Leu-Met-
Clinical use: Topimarate is a sulfamate- Tyr-Asp-Cys-Cys-Thr-Gly-Ser-Cys-Arg-Ser-
substituted derivative of d-fructose with the abil- Gly-Lys-Cys-NH2 cyclic S-3.1-S-3.16:S-
ity to block sodium channels, enhance GABA 3.8-S-3.20:S-3-15-S-3.25-tris(disulfide),
activity by interacting with a nonbenzodiazepine omega-conotoxin MVIIA (reduced),
site of GABAA receptors, and selectively block cyclic (1-16),(8-20),(15-25)-tris(disulfide),
AMPA/kainate glutamate receptors [658]. C102 H172 N36 O32 S7 , M r 2639.14.
The empirical use of topimarate as an Synthesis: Ziconitide is the synthetic form of
antinociceptive drug in humans was established the conotoxin ω-conopeptide MVIIA [675, 676]
before systematic, planned research on animal (see below).
models of pain was initiated. Topiramate was Clinical use: Ziconotide is the synthetic
initially launched in 1995 as monotherapy and equivalent of a naturally occurring conopeptide
adjunctive therapy for the treatment of epilepsy, found in the marine snail Conus magus. Zi-
specifically for the treatment of partial seizures, conotide is an N-type calcium channel blocker,
Lennox–Gastaut syndrome, and primary gen- that was launched in the USA by Elan in 2005 as
eralized tonic clonic seizures in patients inad- a solution for intrathecal infusion for the man-
equately controlled by other anticonvulsants. agement of severe, chronic pain in patients who
Topiramate is also used for the prophylaxis of are refractory to other treatments [675, 676].
migraine headaches. Regulatory approval has been obtained for the
The oral starting dose of topimarate is 25– product in the EU. The drug is administered
50 mg at bedtime and can be increased by 25 through appropriate programmable microinfu-
mg increments weekly to 100–200 mg twice a sion pumps, that can be implanted or external,
day. Main reported side effects include confu- and which release the drug into the fluid sur-
sion, weakness, weight loss, and kidney stones. rounding the spinal cord.
Analgesics and Antipyretics 85

Ziconotide binds to N-type calcium chan- Specific References


nels located on the primary nociceptive (Aγ and 11. J. R. Vane et al., Ann. Rev. Pharmacol. Toxicol.
C) afferent nerves in the superficial layers (rexed 38 (1998) 97 120.
laminae I and II) of the dorsal horn in the spinal 12. K. L. Pierce et al., TIPS 16 (1995) 253 256.
cord. Although the mechanism of action of the 13. C. D. Funk, et al., FASEB J. 5 (1991)
drug has not been established in humans, results 2304–2312.
in animals suggest that blocking of N-type cal- 14. D. A. Kujubu et al., J. Biol. Chem. 266 (1991)
cium channels leads to a blockade of excitatory 12866–72.
15. T. Hla, K. Neilson, Proc. Natl. Acad. Sci. U S
neurotransmitter release from the primary affer-
A. 89 (1992) 7384–8.
ent nerve terminals which induces antinocicep- 16. M. K. O’Banion, et al., Proc. Natl. Acad. Sci.
tion. U S A. 89 (1992) 4888–4892.
In July 2001, ziconotide received orphan drug 17. R. Langenbach et al., Ann. N. Y. Acad. Sci. 889
designation from the EMEA for the treatment (1999) 52–61.
of chronic pain requiring intraspinal analgesia. 18. S. G. Morham et al., Cell 83 (1995) 473–482.
The compound had also been under evaluation 19. J. E. Dinchuk et al., Nature 378 (1995)
at Elan for the treatment of head injury. 406–409.
Intended trade name: Prialt. 20. D. Picot et al., Nature 367 (1994) 243–249.
21. J. R. Kiefer et al., Nature 405 (2000) 97–101.
22. J. R. Vane et al., Annu. Rev. Pharmacol.
4. References Toxicol. 38 (1998) 97–120.
23. J. Berg et al., J. Pharmacol. Toxicol. Meth. 37
General References (1997) 179–186.
1. J. G. Hardman (eds.) et al.: Goodman and 24. A. Graul et al., Drugs Future 22 (1997)
Gilman’s The Pharmacological Basis of 711–714.
Therapeutics, 9th ed., McGraw Hill, New York 25. L. J. Crofford et al., J. Clin. Invest. 93 (1994)
1995, pp. 521–555, 557–600, 617–657. 1095–101.
2. J. E. F. Reynolds (eds.) et al.: Martindale The 26. H. Sawaoka et al., Prostaglandins Leukot.
Extra Pharmacopoeia, 31st ed., The Royal Essent. Fatty Acids 59 (1998) 313–316.
Pharmaceutical Society, London 1996. 27. C. J. McCarthy et al., Am. J. Gastroenterol. 94
3. A. Herz (ed.): Handbook of Experimental (1999) 1218–1223.
Pharmacology, vol. 104/I, Opioids I., vol. 28. R. Langenbach et al., Ann. N.Y. Acad. Sci. 889
104/II, Opioids II, Springer Verlag, Berlin (1999) 52–61.
1993. 29. L. J. Crofford, Curr. Pharm. Des. 17 (2000)
4. T. Dickenson, J. Besson (eds.): Handbook of 1725–36.
Experimental Pharmacology. The 30. C. Bombardier et al., N. Engl. J. Med. 343
Pharmacology of Pain, vol. 130, Springer (2000) 1520–1528.
Verlag, Berlin 1997. 31. R. S. Bresalier et al., N. Engl. J .Med. 352
5. M. Zenz, I. Jurna (eds.): Lehrbuch der (2005) 1092–1102.
Schmerztherapie. Grundlage, Theorie und 32. S. D. Solomon et al., N. Engl. J. Med. 352
Praxis für Aus- und Weiterbildung. (2005) 1071–1180.
Wissenschaftliche Verlagsgesellschaft mbH, 33. N. A. Nussmeier et al., N. Engl. J. Med. 352
Stuttgart 1993. (2005) 1081–1091.
6. J. J. Bonica (ed.): The Management of Pain, 34. B. F. McAdam et al., Proc. Natl. Acad. Sci.
2nd ed., Lea & Febiger, 1990. U.S.A. 96 (1999) 272–277.
7. S. Budavari (ed.): The Merck Index, 12th ed., 35. X. Leval et al., Curr. Med. Chem. 9 (2002)
Merck & Co. Inc., 1996. 941–962.
8. P. D. Wall, R. Melzack (eds.): Textbook of 36. M. Inagaki et al., J. Med. Chem. 43
Pain, 3rd ed., Churchill Livingstone, New (2000)2040-2048.
York 1994. 37. S. Tries et al., Inflamm. Res. 51 (2002)
9. J. R. Vane et al., “Cyclooxygenases 1 and 2,” 135–143.
Annu. Rev. Pharmacol. Toxicol. 38 (1998) 38. S. Laufer et al., Arzneimittelforschung 44
97–120. (1994) 1329–1333.
10. C. J. Hawkey: “Cox-2 Inhibitors”, Lancet 353 39. B. Chin, J. Wallace, Curr. Opin. Investig.
(1999) 307–314. Drugs 1 (1999) 148–152.
86 Analgesics and Antipyretics

40. C. Pisano et al., Dig. Dis. Sci. 44 (1999) 66. D. A. Willoughby et al., Lancet 355 (2000)
713–724. 646–648.
41. S. Fiorucci, E. Antonelli, Curr. Top. Med. 67. P. Kirkpatrick, Nature Reviews Drug
Chem. 5 (2005) 487–492. Discovery 4 (2005) 883.
42. R. Scatena, Curr. Opin. Investig. Drugs 5 68. E. D. Hogestatt et al., J. Biol. Chem. 36 (2005)
(2004) 551–556. 31405–31412.
43. J. L. Wallace et al., Am. J. Physiol. 69. R. K. Lewis, F. P. Paloucek, Clin. Pharm. 10
Gastrointest. Liver Physiol. 286 (2004) (1991) 765–774.
G76–81. 70. M. Depre et al., Fundam. Clin.Pharmacol. 6
44. C. Patrono, N. Engl. J. Med. 330 (1994) (1992) 259–262.
1287–1294. 71. U. C. Dubach et al., N. Engl. J. Med. 308
45. Patrignani et al., Br. J. Pharmacol. 118 (1996) (1983) 357–362.
1285–1293. 72. K. H. Boltze, H. Kreisfeld,
46. R. G. Kurumbail et al., Nature 384 (1996) Arzneim.-Forsch./Drug Res. 27 (1977) no. 1,
644–648. 1300–1312.
47. A. S. Kalgutkar et al. Science 280 (1998) 73. R. Coletta et al., Int. J. Clin. Pharmacol. Res. 8
1268–1270. (1988) 295–298.
48. A. Kleemann, J. Engel, B. Kutscher, D. 74. Parke Davis, FR 1 341 M, 1961.
75. R. B. Moffett, B. D. Aspergen, J. Am. Chem.
Reichert: Pharmaceutical Substances,
Soc. 82 (1960) 1605.
Synthesis, Patents, Applications, Thieme,
76. S. Ravi et al., Postgrad. Med. J. 62 (1986)
Stuttgart – New York,
773–776.
1999 (available in print and CD-ROM).
77. H. Gögelein et al., FEBS Lett. 268 (1990)
49. N. Kuhnert, Chem. Unserer Zeit 33 (1999)
79–82.
213–220.
78. M. M. White et al., Mol. Pharmacol. 37 (1990)
50. Am. J. Med. 74 (1983) no. 6A, 1–109.
720–724.
51. I. Tegeder et al., FASEB J. 15 (2001) 79. L. Sorbera et al., Drugs of the Future 26
2057–2072. (2001) 133–140.
52. C. J. Needs, P. M. Brooks, Clin. 80. G. J. Grover et al., J. Pharmacol. Exp. Ther.
Pharmacokinet. 10 (1985) 164–177. 269 (1994) 536–540.
53. R. J. Waldmann et al., JAMA 247 (1982) 81. Y. T. Lee, Q. Wang, Eur. J. Pharmacol. 378
3089–3094. (1999) 349–356.
54. Merck & Co., US 4 225 730, 1980, (H. Jones, 82. Parke Davis, US 3 313 848, 1967;
R. Houser. DE 1 149 015, 1961.
55. For alternative synthesis: US 3 992 495, 83. P. F. Juby et al., J. Med. Chem. 11 (1968)
(Merck & Co.). 111–117.
56. For alternative synthesis: US 4 131 618, 84. E. Arrigoni-Martelli, Drugs Future 3 (1978)
(Merck & Co.). 307–310.
57. J. Hannah et al., J. Med. Chem. 21 (1978) 85. J. R. McLean, M. I. Geuckman,
1093. Arzneimittel-Forsch. 33 (1983) 627–631.
58. E. Arrigoni-Martelli, Drugs Future 3 (1978) 86. A. Peretz et al. Mol. Pharmacol. 67 (2005)
28–33. 1053–1066.
59. J. Hannah et al., J. Med. Chem. 21 (1978) 87. Y. T. Lee, Q. Wang, Eur. J. Pharmacol. 378
1093–1100. (1999) 349–356.
60. M. Cotton, R. A. Hux, Anal. Profiles Drug 88. Parke Davis, US 3 138 636, 1964;
Subst. 14 (1985) 491–526. DE 1 163 846, 1961.
61. J.M. Young et al., Inflamm Res. 45 (1996) 89. G. R. Singh, P. Choudhury, Indian Pediatr. 26
246–253. (1989) 577–579.
62. Lunbeck, GB 656 746, 1948. 90. M. Ottolia, L. Toro, Biophys. J. 67 (1994)
63. D. S. Hoffenberg, C. R. Hauser, J. Org. Chem. 2272–2279.
20 (1955) 1496–1500. 91. J. S. Marks, M. H. Gleeson, Br. Med. J. 4
64. Warner-Lambert, US 2 998 450, 1961, (G. (1975) 442.
Wilbert, J. De Angelis). 92. Labs. U.P.S.A., US 3 415 834, 1968;
65. B. Ameer et al., Ann. Int. Med. 87 (1977) US 3 337 570, 1967.
93. J. Auclair et al., Curr. Ther. Res. 46 (1989)
202–209.
782–788.
Analgesics and Antipyretics 87

94. M. Civelli et al., Agents Actions 33 (1991) 123. J. R. Carson et al., J. Med. Chem. 14 (1971)
233–239. 646–647.
95. Park Davis, US 3313848 (A. Scherer, F. W. 124. R. N. Brogden, Drugs 15 (1978) 429–450.
Short), 1967 125. C. Brideau et al., Inflamm Res. 45 (1996)
96. M. Zovko, B. Zorc, M. J.-M. Takac, B. 68–74.
Metelko, P. Novac, Croatica Chemica Acta 76 126. E. Arrigoni-Martelli, J. Prous, J. Castaner,
(2003), 335–341. Drugs of the Future 14 (1989) 963.
97. T. Corell, Pharmacol. Toxicol. 75 Suppl 2 127. G. Biasi, R. Marcolongo, Minerva Med. 92
(1994) 14–21. (2001) 315–324.
98. H. Isomaki, Pharmacol. Toxicol. 75 Suppl 2 128. G. Coruzzi et al., Curr. Drug Targets Inflamm.
(1994) 64–65. Allergy. 3 (2004) 43–61.
99. J. Delgado et al. Pharmacol. Toxicol. 75 Suppl 129. G. Coruzzi et al., Dig. Liver Dis. 34 (2002)
2 (1994) 89–91. 403–410.
100. P. E. Hansen, Pharmacol. Toxicol. 75 Suppl 2 130. J. Prous, J. Castaner, Drugs of the Future 13
(1994) 81–82. (1988) 1050.
101. K. H. Boltze et al., Arzneim.-Forsch./Drug 131. CDC Life Sciences Inc., US 4 327 222, 1982
Res. 30 (1977) no. 2 1314–1325. (R. G. Micetich et al.).
132. Teiho Pharmaceuticals, EP 454 871, 1990 (H.
102. H. Jacobi, H. D. Dell, Arzneimittel-Forsch. 30
Tanaka, Y. Hagiwara, H. Kajitani, H.
(1980) 1348–1362.
Yasumoto).
103. Rotta Research Lab., DOS 2 535 799, 1975.
133. Taiho Pharmaceuticals, EP 464 218, 1991 (H.
104. Rotta Research Lab., US 3 985 878, 1974.
Tanaka, Y. Hagiwara, H. Kajitani, H.
105. F. Makovec et al., Arzneimittelforschung 37
Yasumoto).
(1987) 806–813.
134. CDC Life Science, EP 26928, 1981 (R. G.
106. V. Fossaluzza et al., Minerva Med. 74 (1983)
Micetich et al.).
723–726. 135. T. Ushio et al, Iykuhin Kenkyu 22 (1991) 892.
107. N. P. Buu-Hoi et al., Hebd. Seances Acad. Sci. 136. N. Ono et al., Nippon Yakurigaku Zasshi. 95
261 (1965) 2259. (1990) 63–81.
108. P. Moser et al., J. Med. Chem. 33 (1990) 137. K. Goto et al., Prostaglandins Other Lipid
2358–2368. Link Mediat. 56 (1998) 245–254.
109. P. A. Todd, E. M. Sorkin, Drugs 35 (1988) 138. D. A. Walch et al., J. Med. Chem. 27 (1984)
244–285. 1379–1388.
110. T. Y. Shen et al., J. Am. Chem. Soc. 85 (1963) 139. US 4 126 635, 1977 (H. Robins et al.).
488–489. 140. A.H. Robinson Co., Inc. US 4 568 695, 1983
111. Merck & Co., US 3 161 654, 1964, (H. Robins et al.).
DE 1 232 150, 1962. 141. J. Prous, J. Castaner, Drugs of the Future 13
112. H. Yamamoto et al., Chem. Pharm Bull. 16 (1988) 943.
(1968) no. 17, 647. 142. T. Ogawa et al. Nippon Ganka Gakkai Zasshi
113. Byk Gulden, US 4 4 325 962, 1982; 99 (1995) 406–411.
DE 1 946 370, 1969. 143. R. J. Fontana et al., Liver Transpl. Surg. 5
114. G. Rainer et al., Arzneim.-Forsch./Drug Res. (1999) 480–484.
31 (1981) 649–655. 144. E. B. Hunter et al., Am. J. Gastroenterol. 94
115. B. Unterhalt, Drugs Future 7 (1982) 110–111. (1999) 2299–2301.
116. Merck & Co., US 3 882 239, 1975; 145. P. L. Moses et al., Am. J. Gastroenterol. 94
DE 1 039 426, 1970. (1999) 1393–1396.
117. R. F. Shuman et al., J. Org. Chem. 42 (1977) 146. Beecham, US 4 420 639, 1983;
1914–1919. DE 2 463 219, 1974.
118. R. N. Brogden et al., Drugs 16 (1987) 97–114. 147. A. C. Goudie et al., J. Med. Chem. 12 (1978)
119. A. Whelton et al., JAMA 249 (1983) 2892. 1260–1264.
120. T. D. Warner et al., Proc. Natl. Acad. Sci. 148. M. Neumann, Drugs Future 6 (1981) 35–36.
U.S.A. 96 (1999) 7563–7568. 149. C. Prabhakar et al., Org. Process Res. Dev. 3
121. C. Haanen, Curr. Opin. Investig. Drugs 2 (1999) 121–125.
150. H. A. Friedel et al., Drugs 45 (1993) 131–156.
(2001) 677–683
151. Degussa, US 3 513 171, 1970;
122. McNeil, US 3 752 826, 1973;
DE 1 670 522, 1966.
GB 1 428 272, 1973. 152. Drugs Future 8 (1983) 773–775.
88 Analgesics and Antipyretics

153. W. v. Bebenburg, et al., Chem. Ztg. 105 (1981) 183. J. Pirotte et al., Gastroenterol. Clin. Biol. 11
217–219. (1987) 520.
154. W. v. Bebenburg, G. Steinmetz, K. Thiele, 184. J. R. Hannequin et al., Rev. Rheum. Mal.
Chem. Ztg. 103 (1979) 387–399. Osteoartic. 55 (1988) 983–988.
155. M. L. Brochier, Eur. Heart J. 14 (1993) 185. A. C. Depla et al., Neth. J. Med. 37 (1990)
951–957. 32–36.
156. W. J. Wechter et al., Cancer Res. 60 (2000) 186. M. Eugene et al., Rev. Med. Interna 8 (1987)
2203–2208. 115.
157. J. M. Mayer, B. Testa, Drugs Future 22 (1997) 187. Ely Lilly, DOS 1 941 625, 1969.
1347–1366. 188. Ely Lilly, US 3 600 437, 1971 (W. S.
158. M. Cleij et al., J. Org. Chem. 64 (1999) Marshall).
5029–5035. 189. Mitsubishi Petrochemical, DOS 2 646 792,
159. M. Busson, J. Int. Med. Res. 14 (1986) 53–62. 1976 (M. Takeda, M. Uchide, H. Masayuka).
160. Rhône-Poulence, US 3 641 127, 1972. 190. Nissin Flour Milling, US 4 016 196, 1977 (K.
161. G. G. Liversidge, Anal. Profiles Drug Subst. Katsura Kogure, T. Noriyoshi Sueda, T.
10 (1981) 443–471. Youziro Yoshino, K. Kunio Nakagawa).
162. J. L. Hommeril et al., Br. J. Anaesth. 72 (1994) 191. Sagami, DAS 2 709 504, 1977 (G.
383–387. Tsuchihashi, K. Ogura).
163. P. Patrignani et al., J Physiol. Pharmacol. 48 192. C. M. Gruber Jr., J. Rheumatol. 2 (1976) 8–17.
(1997) 623–631. 193. R. N. Brogden et al., Drugs 13 (1977)
164. R. L. Dorfman, Arzneim.-Forsch./Drug Res. 25 241–265.
(1975) 278–281. 194. R. N. Brogden et al., Drugs 21 (1981) 1–22.
165. T. Y. Shen, Angew. Chem. Int. Ed. Engl. 11 195. J. R. Ryan et al., Clin. Pharmacol. Ther. 42
(1972) 460. (1987) 28–32.
166. H. R. Bellur, J. R. Ahuja, D. G. Kulkarni, 196. M. L. Wendland et al., Mayo Clin. Proc. 55
Tetrahedron: Asymmetry 3 (1992) 163–192. (1980) 103–107.
167. Syntex, US 3 663 584, 1972. 197. J. S. Stotts et al., J. Am. Acad. Dermatol. 18(4
168. Syntex, US 4 605 758, 1986. Pt 1) (1988) 755–757.
169. P. A. Todd, S. P. Clissold, Drugs 40 (1990) 198. CERM, US 4 218 473, 1980 (J. J. Godfroid, E.
91–137. Steiner).
170. J. Rautio et al., J. Med. Chem. 43 (2000) 199. M. Hoeijmakers et al., J. Vet. Pharmacol. Ther.
1489–1494. 28 (2005) 305–312.
171. Roussel-Uclaf, DOS 2 055 264, 1970. 200. T. Nell et al., J. Small Anim. Pract. 43 (2002)
172. G. L. Plosker, A. J. Wagstaff, Drugs 50 (1995) 208–212.
1050–1075. 201. P. Lees et al., J. Vet. Pharmacol. Ther. 22
173. F. G. Mayall et al., Br. Med. J. 309 (1994) (1999) 96–106.
599–600. 202. D. A. Walsh et al., J. Med. Chem. 33 (1990),
174. J. Prous, J. Castaner, Drugs of the Future 14 2296–2304.
(1989) 302. 203. Alcon, WO 09932104, 2000 (J. H. Yanni, M.
175. Nippon Chemiphar, US 4 247 706, 1981 (Y. R. Hellberg).
Fujimoto, S. Yamabe). 204. T. L. Ke et al., Inflammation 24 (2000)
176. M. Hatori, S. Kokubun, Curr. Med. Res. Opin. 371–384.
14 (1998) 79–87. 205. K. Takahashi et al., Invest. Ophthalmol. Vis.
177. N. Murakami et al., Jpn. J. Pharmacol. 72 Sci. 44 (2003) 409–415.
(1996) 29–37. 206. D. A. Gamache et al., Inflammation 24 (2000)
178. H. B. Tang et al., Neuropharmacology 48 357.
(2005) 1035–1042. 207. R. Linstrom, T. Kim, Curr. Med. Res. Opin. 22
179. R. W. Carney et al., Experientia 29 (1973), (2006) 397–404.
938. 208. W. J. Welstead et al., J. Med. Chem. 22 (1979),
180. Ciba, US 3 641 040, 1972 (R. W. Carney, G. 1074–1079.
DeStevens). 209. Robins Co., Inc., DE 2 324 768, 1973 (W.
181. P. A. Todd, R. Beresford, Drugs. 32 (1986) Welstead, H. W. Moran)
509–537. 210. Robins Co., Inc., US 1972 254 285, 1972 (W.
182. M. Guidotti et al., J. Int. Med Res. 17 (1989)
Welstead, H. W. Moran)
48–54.
Analgesics and Antipyretics 89

211. G. Ehrhart, H. Ruschig: Arzneimittel, 243. Hoffmann-La Roche, DOS 2 537 070, 1975.
Entwicklung, Wirkung, Darstellung, vol. 1, 244. R. N. Brogden et al., Drugs 22 (1981)
Verlag Chemie, Weinheim 1972, pp. 171–172. 165–187.
212. I.G. Farben, DRP 476 663, 1922. 245. R. N. Brogden et al., Drugs 28 (1984)
213. Z. Fendrich, Cas. Lek. Cesk. 139 (2000) 292–323.
440–444. 246. J. A. Mitchell et al., Proc. Natl. Acad. Sci. USA
214. F. Arellano, J A. Sacristan, Eur. J. Clin. 90 (1993) 11693–11697.
Pharmacol. 38 (1990) 617–619. 247. E. Arrigoni-Martelli, Drugs Future 7 (1982)
215. M. Levy et al., Clin. Pharmacokinet. 28 (1995) 493–494.
216–234. 248. P. A. Todd, S. P. Clissold, Drugs 41 (1991)
216. Hoffmann-La Roche, DRP 565 799, 1931. 625–646.
217. Riedel-deHaen, DE 962 254, 1954. 249. Synthex, US 4 347 186, 1982;
218. K. Brune, Agents Actions 25 Suppl. (1988) US 4 458 081, 1984;
9–19. US 4 873 340, 1989.
219. E. Gores et al., Med. Monatsschr. Pharm. 27 250. E. Arrigoni-Martelli, Drugs Future 8 (1983)
(2004) 72–76. 871–874.
220. R. Denss et al., Helv. Chim. Acta 40 (1957) 251. A. Guzman et al., J. Med. Chem. 29 (1986)
402. 589–591.
221. M. Kühn et al., J. Prakt. Chem. 156 (1940) 252. J. M. Muchowski et al., J. Med. Chem. 28
103. (1985) 1037–1049.
222. Horakova et al., Pharmacotherapeutica (1963) 253. F. F. Franco et al., J. Org. Chem. 47 (1982)
335–350. 1682–1688.
223. J. Büchi et al., Helv. Chim. Acta 36 (1953) 75. 254. M. M. T. Buckley, R. N. Brogden, Drugs 39
224. Comm. Farmaceutica Milanese, GB 839 057, (1990) 86–109.
1957. 255. John Wyeth and Brothers, Ltd, US 3578671,
225. D. Loew et al., Z. Rheumatol. 44 (1985) 1971 (K. Brown).
186–192. 256. E. Arrigoni-Martelli, Drugs of the Future 3
226. Geigi, US 2 745 783, 1956. (1978) 539–542.
227. L. E. Westerholm, Br. Med. J. 3 (1973) 257. L. G. Miller, Clin. Pharm. 11 (1992) 591–603.
339–343. 258. American Home Products, US 3 843 681,
228. Geigi, US 2 562 830, 1951. 1974.
229. S. L. Ali, Anal. Profiles Drug Subs. 11 (1982) 259. L. Castaner, E. Arrigoni-Martelli, Drugs
483–521. Future 2 (1977) 21–23.
230. G. A. Faich, Pharmacotherapy 7 (1987) 260. C. A. Demerson, et al., J. Med. Chem. 19
25–27. (1976) 391–395.
231. Warner-Lambert, DOS 2 208 351, 1972. 261. C. A. Demerson et al., J. Med. Chem. 18
232. J. G. Lombardino, E. H. Wisemann, J. Med. (1975) 189.
Chem. 14 (1971) 973. 262. N. Bellamy, Inflammopharmacology 5 (1997)
233. H. Zinnes et al., J. Med. Chem. 25 (1982) 12. 139–152.
234. W. W. Downie, Semin. Arthritis Rheum. 12 263. A. S. Taha et al., Gut 30 (1989) A751.
(1982) Suppl. 2, 170–174. 264. G. Bianchi Porro, et al., J. Intern. Med. 229
235. Drugs Future 17 (1992) 683–686.
(1991) 5–8.
236. T. Pruss et al., Postgrad. Med. J. 66 (1990)
265. Thomae GmbH, DE 2 756 113, 1979.
Suppl. 4818–821.
266. G. Trummlitz, G. Engelhardt, U. Busch, Drugs
237. T. Christoph et al., Inflamm. Res. 44 (1995)
Future 14 (1989) 1047–1048.
Suppl. 3 Abstr. W16/17.
267. G. Engelhardt et al., Inflamm. Res. 44 (1995)
238. J. A. Balfour et al., Drugs 51 (1996) 639–657.
423–433.
239. Pfizer, US 3 591 584, 1971;
268. J. A. Lopez-Garcia et al., Neuroreport 9
US 4 469 866, 1984.
(1998) 647–651.
240. J. G. Lombardino et al., J. Chiaini, J. Med.
269. Riker, US 3 840 597, 1974;
Chem. 5 (1973) 493–496.
DOS 2 333 643, 1973.
241. E. H. Wisemann et al., Arzneim.-Forsch./Drug
270. R. Davis, R. N. Brogden, Drugs 48 (1994)
Res. 26 (1976) 1300–1303.
431–454.
242. L. Castaner, E. Arrigoni-Martelli, Drugs
271. A. Azab et al., Life Sci. 63 (1998) PL 323–327.
Future 2 (1977) 124–127.
90 Analgesics and Antipyretics

272. P. A. McCormick et al., Lancet 353 (1999) 299. E. Packman et al., Headache 45 (2005)
40–41. 1163–1170.
273. C. Ferreiro et al., Gastroenterol. Hepatol. 23 300. J. A. Rosenberg et al., Nat. Clin. Pract.
(2000) 428–430. Gastroenterol. Hepatol. 2 (2005) 14–15.
274. Drugs Future 22 (1997) 711–714. 301. C. J. Hawkey et al., Clin. Gastroenterol.
275. Searle & Co., US 5 521 207; EP 731 795; WO Hepatol. 4 (2006) 57–66.
9 515 316 (J. J. Talley, T. D. Penning, P. W. 302. P. Matchaba et al., Clin. Ther. 27 (2005)
Collins et al.). 1196–214.
276. G. Steinbach et al., N. Engl. J. Med. 342 303. H. Tannenbaum et al., Ann. Rheum. Dis. 63
(2000) 1946–1952. (2004) 1419–1426.
277. J. L. Goldstein et al., Am. J. Gastroenterol. 96 304. C. M. Rordorf et al., Clin. Pharmacokinet. 44
(2001) 1019–1027 (2005) 1247–1266.
278. F. E. Silverstein et al., JAMA 284 (2000) 305. Pfizer Inc., WO 2005123701, 2005 (L. J.
1247–1255. Letendre, C. K. Snoddy, G. H. Klemm, J. P.
279. J. M. Brophy, Expert Opin. Drug. Saf. 4 (2005) Lawson, M. J. Bauer).
1005–1015. 306. J. J. Talley et al., J. Med. Chem. 43 (2000)
280. L.A. Sorbera et al., Drugs of the Future 26 1661–1663.
(2001) 346–353. 307. Searle & Co. WO 9515316;
281. R.W. Friesen et al., Biorg. Med. Chem. Lett. 8 US 5 521 207;
(1999) 2777–2782. EP 731 795 1995 (J. J -Talley et al.).
282. I. W. Davies et al., J. Org. Chem. 65 (2000) 308. S. M. Cheer, K. L. Goa, Drugs 61(2001)
8415–8420. 1133–1141.
283. I. W. Davies et al., J. Org. Chem. 65 (2000) 309. A. W Gotta, Idrugs 4 (2001) 939–944.
8571–8574. 310. A. S. Dalpiaz, D. Peterson, Expert. Rev.
284. J. F. Marcoux et al., Org. Lett. 2 (2000) Neurother. 4 (2004) 165–177.
2339–2341. 311. S. Aldington et al., N. Z. Med. J. 118 (2005)
285. A. Dallob et al., J Clin Pharmacol. 43 (2003) U1755.
573–585. 312. C. D. Furberg et al., Circulation 111 (2005)
286. D. J. Cochrane et al., Drugs. 62 (2002) 249.
2637–2651. 313. E. G. Harks et al., J. Pharmacol. Exp. Ther.
287. A. D. Rodrigues et al., Drug Metab. Dispos. 31 298 (2001) 1033–1041.
(2003) 224–232. 314. L. A. Sorbera et al. Drugs Future 23 (1998)
288. K. Kassahun et al., Drug Metab. Dispos. 29 1287–1296.
(2001) 813–820. 315. L. A. Sorbera et al., Drugs of the Future 23
289. N. Chauret et al., Bioorg. Med. Chem. Lett. 11 (1998) 1287–1296.
(2001) 1059–1062. 316. M. Therien et al., Synthesis (2001) 1778–1779.
290. D. R. Ramey et al., Curr. Med. Res. Opin. 21 317. L. J. Scott, H. M. Lamb Drugs. 58 (1999)
(2005) 715–722. 499–505.
291. S. P. Curtis et al., Clin Ther. 26 (2004) 70–83. 318. A. J. Matheson, D. P. Figgitt, Drugs. 61 (2001)
292. S. Aldington et al., N. Z. Med. J. 118 (2005) 833–865.
U1684. 319. C. C. Chan et al., J. Pharmacol. Exp. Ther. 290
293. L. A. Sorbera et al., Drugs of the Future 27 (1999) 551–560.
320. L. Laine et al., Gastroenterology 117 (1999)
(2002) 740–747.
776–783.
294. Novartis, WO 2001 023346, 2001 (M. Murat,
321. C. Bombardier et al., N. Engl. J. Med. 343
T. Allemendiger, J. Calienni, J. Cercus, O.
(2000) 1520–1528.
Loiseleur).
322. M. Wadman, Nature 433 (2005) 790.
295. Novartis, WO 1999 011605, 1999 (L.
323. J. J. Talley in F. D. King A. W. Oxford (eds.):
Mcquire, B. Mugrage, J. Van Duzer).
Progress in Medicinal Chemistry-Vol 36,
296. K. A. Lyseng-Williamson et al., Drugs 64
Elsevier, Amsterdam 1999.
(2004) 2237–2246.
324. D. Ormrod et al., Drugs 62 (2002) 2059–2071.
297. F. Berenbaum et al., J. Int. Med. Res. 33
325. M. L. Chavez, C. J. De Korte Clin Ther. 25
(2005) 21–41.
(2003) 817–851.
298. P. Geusens et al., Int. J. Clin. Pract. 58 (2004)
326. C. Fenton et al. Drugs. 64 (2004)1231–1261.
1033–1041.
327. J. E. Edwards et al. Pain 111 (2004) 286–296.
Analgesics and Antipyretics 91

328. G. P.Joshi Expert Rev. Neurother. 5 (2005) 353. R. J. Knapp et al., FASEB J. 9 (1995) 516–525.
11–24. 354. S. R. Childers Life Sci. 48 (1991) 1991–2003.
329. D. Kudrow et al. Headache 45 (2005) 355. I. McFadzean, Neuropeptides 11 (1988)
1151–1162. 173–180.
330. C. Talhari et al., J. Allergy Clin. Immunol. 115 356. W. F. Simonds, Endocr. Rev. 9 (1988) 200–212.
(2005) 1089–1090. 357. A. D. Blake et al. Channels 5 (1997) 231–235.
331. F. L. Byerly et al., Burns 31 (2005) 383–387. 358. P. Y. Law, H. H. Loh, J. Pharmacol. Exp. Ther.
332. N. A. Nussmeier et al. N. Engl. J. Med. 352 289 (1999) 607–624.
(2005) 1081–1091. 359. N. I. Cherny, Drugs 51 (1996) 713–737.
333. A. F. Casy, R. T. Parfitt: Opioid Analgesics, 360. H. W. Matthes et al., Nature 383 (1996)
Chemistry and Receptors, Plenum Press, New 819–823.
York 1986. 361. B. L. Kieffer, Trends Pharmacol. Sci. 20
334. A. F. Casy, R. T. Parfitt, Opioid Analgesics. (1999) 19–26.
Chemistry and Receptors, Plenum Press, New 362. D. I. Scopes, Drug of the Future 18 (1993)
York -London 1986. 933–947.
335. A. F. Casy, R. T. Parfitt, Opioid Analgesics. 363. A. Barber, R. Gottschlich, Exp. Opin. Invest.
Chemistry and Receptors, Plenum Press, New Drugs 6 (1997) 1354–1368.
York -London 1986. 364. G. Dondio et al., Exp. Opin. Ther. Patents 7
336. H. Akil et al. Annu. Rev. Neurosci. 7 (1984) (1997) 1075–1098.
223–255. 365. M. A. Scheideler, Curr. Opin. CPNS Invest
337. A. Mansour et al., Trends Neurosci. 18 (1995) Drugs 2 (2000) 171–177.
22–29. 366. C. Mollereau, L. Mouledous, Peptides. 21
338. A. Herz J. Neural. Transm. Suppl. 18 (1983) (2000) 907–917.
227–233. 367. R. Bertorelli et al., Trends Pharmacol. Sci. 21
339. G. B. Stefano, et al. Crit. Rev. Immunol. 16 (2000) 233–234.
(1996) 109–144. 368. A. Mansour et al., Trends Neurosci. 18 (1995)
340. C. Stein et al.,in C. Stein (ed.): Opioids in Pain 22–29.
Control. Basic and Clinical Aspects, 369. J. Lipp, Clin. Neuropharmacol. 14 (1991)
Cambridge University Press, Cambridge 1999, 131–147.
pp. 96–108. 370. T. L Yaksh, Acta Anaesthesiol. Scand. 41
341. W.R. Martin et al., J. Pharmacol. Exp Ther. (1997) 94–111.
197 (1976) 517–5132. 371. C. Stein, Opioids in Pain Control. Basic and
342. C. J. Fowler G. L. Fraser Neurochem. Int. 24 Clinical Aspects, Cambridge University Press,
(1994) 401–426. Cambridge 1999.
343. W. R. Martin, Pharmacol. Rev. 35 (1983) 372. C. P. Watson, Clin. J. Pain, 16 (2 Suppl.)
283–323. (2000) S49–55.
344. B. N. Dhawan et al., Pharmacol Rev. 48 373. R. C. Etches et al., Can. J. Anaesth. 36 (1989)
(1996) 567–592. 165–185.
345. J. C. Meunier et al., Nature 377 (1995) 374. J. E. Shook et al., Am. Rev. Respir. Dis. 142
532–535. (1990) 895–909.
346. P. L. Wood, Neuropharmacology 21 (1982) 375. M. F. Mulroy, Reg. Anesth. 21 (6 Suppl.)
487–497. (1996) 89–93.
347. G. W. Pasternak, P. J. Wood, Life Sci. 38 376. F. W. Jackson, Gastroenterology 106 (1994)
(1986) 1889–1898. 820–821.
348. G. Gaveriaux-Ruff, B. Kieffer, in C. Stein, 377. T. A. Bowdle, Drug Saf. 19 (1998) 173–189.
(ed.): Opioids in Pain Control. Basic and 378. W. Kromer, Dig. Dis. 8 (1990) 361–373.
Clinical Aspects, Cambridge University Press, 379. I. Mancini, E. Bruera, Support Care Cancer 6
Cambridge 1999, pp. 1–20. (1998) 356–364.
349. B. L. Kieffer et al., Proc. Natl. Acad. Sci. U S 380. A. De Luca, I. M. Coupar, Pharmacol. Ther.
A. 89 (1992) 12048–12052. 69 (1996) 103–115.
350. C. J. Evans et al., Science, 258 (1992) 381. R. C. Heel et al. Drugs. 15 (1978) 33–52.
1952–1955. 382. E. Campora et al., J. Pain Symptom Manage. 6
351. R. K. Reinscheid et al. Science, 270 (1995) (1991) 428–430.
792–794. 383. R. Aparasu et al., J. Pain Symptom Manage. 18
352. G. Calo’ et al. Peptides, 21 (2000) 935–947. (1999) 280–288.
92 Analgesics and Antipyretics

384. J. P. Blancquaert et al. Eur. J. Pharmacol. 128 412. V. Ozdemir et al., Pharmacogenetics 10
(1986) 143–150. (2000) 373–388.
385. N. M. Barnes et al., Neuropharmacology 30 413. G. Ehrhart, H. Ruschig: Arzneimittel,
(1991) 1073–1083. Entwicklung, Wirkung, Darstellung, vol. 1,
386. A. Pfeiffer et al., Science 233 (1986) 774–776. Verlag Chemie, Weinheim 1972, pp. 79, 115.
387. R. Brown, R. Lo, Aust. N. Z. .J Ment. Health 414. C.E. Inturrisi et al., Life Sci. 33 Suppl 1 (1983)
Nurs. 9 (2000) 65–74. 773–776.
388. D. A. Taylor, W. W. Fleming, Pharmacol. Exp. 415. J. G. Umans, C. E. Inturrisi, J. Pharmacol.
Ther. 297 (2001) 11–18. Exp. Ther. 218 (1981) 409–415.
389. H. A. Heit, Eur. J. Pain 5 Suppl A (2001) 416. D. K. Wyatt, L. T. Grady, Anal. Profiles Drug
27–29. Subst. 10 (1981) 357–403.
390. R. F. Reder, Eur. J. Pain. 5 Suppl A (2001) 417. U. Boerner et al., Drug Metab. Rev. 4 (1975)
109–111. 39–73.
391. F. J. Muhtadi, Anal. Profiles Drug Subs. 17 418. C. E. Inturrisi et al., N. Engl. J. Med. 310
(1988) 259–366. (1984) 1213–1217.
392. Knoll, DOS 2 726 925, 1977. 419. G. Ehrhart, H. Ruschig: Arzneimittel,
393. T. Hudlicky et al.: “A historical perspective of Entwicklung, Wirkung, Darstellung, vol. 1,
morphine synthesis,” in: Atta-ur-Rahman Verlag Chemie, Weinheim 1972, p. 180.
(ed.): Studies in Natural products Chemistry, 420. A. Stein, Pharmazie 10 (1955) 180–186.
Elsevier, Amsterdam 1996, pp. 43–116. 421. J. E. Edwards et al. Cochrane Database Syst.
394. D. Trauner et al., Synthesis 1983 203–204. Rev. (2000) CD 2760.
395. J. Mulzer, D. Trauner, Chirality 11 (1999) 422. H. Matthys et al., Schweiz. Med. Wochenschr.
475–482. 115 (1985) 307–311.
396. J. Frackenpohl, Chem. u. Zeit 34 (2000) 423. R. S. Lloyd et al., Curr. Med. Res. Opin. 13
99–112. (1992) 37–48.
397. G. J. Meuzelaar et al., Eur. J. Org. Chem. 424. Reckitt & Colman, DE 1 620 206, 1966.
(1999) 2315–2321. 425. Reckitt & Sons Ltd., US 3 433 791, 1969.
398. J. Mulzer, D. Trauner, Chirality 11 (1999) 426. K. W. Bentley, D. G. Hardy, Proc. Chem. Soc.
475–482. (London) 1963, 220.
399. H. Nagata, et al. Chem. Commun. (2001) 427. W. G. Dorner, Chem. Unserer Zeit 15 (1986)
1094–1095. 33–46.
400. H. J. McQuay, B. J. Anaest. 63 (1989) 428. K. O. Lee et al., Eur. J. Pharmacol. 378 (1999)
213–226. 323–330.
401. L L. Christrup, Acta Anaesthesiol. Scand. 41 429. G. F. Blane, J. Pharm. Pharmacol. 19 (1967)
(1997) 116–122. 367–373.
402. R. Osborne et al., Clin. Pharmacol. Ther. 47 430. T. Alford et al., J. Am. Vet. Med. Ass. 164
(1990) 12–19. (1974) 702–705.
403. J. Lotsch, G. Geisslinger, Clin. 431. G. Ehrhart, H. Ruschig: Arzneimittel,
Pharmacokinet. 40 (2001) 485–499. Entwicklung, Wirkung, Darstellung, vol. 1,
404. G. Ehrhart, H. Ruschig: Arzneimittel, Verlag Chemie, Weinheim 1972, pp. 119–120.
Entwicklung, Wirkung, Darstellung, vol. 1, 432. Knoll, DRP 607 931, 1935.
Verlag Chemie, Weinheim 1972, pp. 117–118. 433. Z. R. Chen et al., Life Sci. 48 (1991)
405. Boehringer, DRP 247 180, 1912. 2165–2171.
406. B. L. Posey, N. S. Kimble, J. Anal. Toxicol. 8 434. A. B. Morrison, Can. Med. Assoc J. 120
(1984) 68–74. (1979) 1338.
407. F. J. Muhtadi, M. M. A. Hassan, Anal. Profiles 435. Merck, US 2 715 626, 1955.
Drug Subs. 10 (1981) 93–138. 436. H. Rapoport, R. Naumann, E. R. Bissel, R. M.
408. Q. Y. Yue et al., Ther. Drug Monit. 19 (1997) Bonner, J. Org. Chem. 15 (1950) 1103–1107.
539–542. 437. G. Lindena et al., Schmerz 12 (1998) 195–204.
409. G. Ehrhart, H. Ruschig: Arzneimittel, 438. D. E. Moulin et al., Lancet 337 (1991)
Entwicklung, Wirkung, Darstellung, vol. 1, 465–468.
Verlag Chemie, Weinheim 1972, 118. 439. J. L. Hill, J. P. Zacny, Psychopharmacology
410. E. Merck AG, DRP 131 980, 1902.
(Berlin). 152 (2000) 31–39.
411. E. Tani et al., Arzneimittelforschung 39 (1989)
1399–1402.
Analgesics and Antipyretics 93

440. G. Ehrhart, H. Ruschig: Arzneimittel, 470. C. B. Christensen, Pharmacol. Toxicol. 73


Entwicklung, Wirkung, Darstellung, vol. 1, (1993) 344–345.
Verlag Chemie, Weinheim 1972, pp. 130–131. 471. L. Jylli et al., Acta Anaesthesiol. Scand. 48
441. O. Schnider, A. Grüssner, Helv. Chim. Acta 34 (2004) 1256–1259.
(1951) 2211–2217. 472. S. Andersen et al., Pain 67 (1996) 369–374.
442. O. Schnider, J. Hellerbach, Helv. Chim. Acta 473. P. Peltola, P. Soisalo, AMA Arch Intern Med.
33 (1950) 1437–1448. 101 (1958) 741–746.
443. R. Grewe, Naturwissenschaften 33 (1946) 474. Janssen, US 3 164 600, 1965;
333–337. US 3 141 823, 1964.
444. S. R. Childers, Life Sci. 48 (1991) 1991–2003. 475. R. Hess et al., J. Pharmacol. Exp. Ther. 179
445. L. L. Randall, G. Lehmann, J. Pharmacol. (1971) 474–484.
Exp. Ther. 99 (1950) 163. 476. M. A. Clotz, M. C. Nahata, Clin. Pharm. 10
446. S. W. Coniam, Anaesthesia 46 (1991) 518. (1991) 581–593.
447. G. Ehrhart, H. Ruschig: Arzneimittel, 477. J. F. Buchanan, C. R. Brown, Med. Toxicol. 3
Entwicklung, Wirkung, Darstellung, vol. I, (1988) 1–17.
Verlag Chemie, Weinheim 1972, 118. 478. Janssen, DE-OS 2 819 837, 1978.
448. E. Merck, DRP 411 530, 1925. 479. F. Janssen et al., J. Med. Chem. 29 (1986)
449. K. W. Bentley: The Chemistry of Morphine 2290–2297.
Alkaloids, Oxford 1954. 480. S. J. Hopkins, Drugs Future 6 (1981) 335–337.
450. F. M. Hauser et al., J. Med. Chem. 17 (1974) 481. R. F. Cookson et al., Br. J. Anaesth. 55 Suppl 2
1117. (1983) 147S–155S.
451. J. Cleary et al., J. Pharmacol. Exp. Ther. 271 482. C. J. E. Niemegeers, P. A. J. Janssen, Drug.
(1994) 1528–1534. Dev. Res. 1 (1981) 83.
452. E. Kalzo, A. Vanino, Clin Pharmacol. Ther. 47 483. G. E. Larijani, M. E. Goldberg, Clin. Pharm. 6
(1990) 639–646. (1987) 275–282.
453. F. Kaiko et al., Clin. Pharmacol. Ther. 59 484. Glaxo, EP 383 579, 1990.
(1996) 52–61. 485. Drugs Future 19 (1994) 1088–1092.
454. U. Weiss, J. Am. Chem. Soc. 77 (1955) 486. K. Flick et al., Arzneim.-Forsch./Drug Res. 28
5891–5892. (1978) no. 1, 107–113.
455. R. S. Sinatra, D. M. Harrison, Clin. Pharm. 8 487. W. E. Hoffman et al., Anesthesiology 79
(1989) 541–544. (1993) 107–113.
456. V. S. Copland et al., Am. J. Vet. Res. 48 (1987) 488. T. E. Egan, Clin. Pharmacokinet. 29 (1995)
1626–1630. 80–94.
457. Winthrop, US 2 167 351, 1939, (O. Eisleb). 489. F. Servin, Eur. J. Anaesthesiol. 15
458. I. G. Farben, DE 679 281, 1937. Suppl.(1997) 41–44.
459. O. Eisleb, Chem. Ber. 74 (1941) 1433–1450. 490. C. G. Haigh, Minerva Anesthesiol. 66 (2000)
460. E. E. Smissman, G. Hite, J. Am. Chem. Soc. 81 414–416.
(1959) 1201–1203. 491. J. Cohen, D. l. Royston, Curr. Opin. Crit. Care
461. C. B. Pert, S. H. Snyder, J. Med. Chem. 19 7 (2001) 227–231.
(1976) 1248–1250. 492. L. G. Michelsen et al., J. Clin. Anesth. 8
462. N. P. Fish, N. J. De-Angelis, Anal. Profiles (1996) 679–682.
Drug Subs. 1 (1972) 175–205. 493. Janssen, US 3 998 834, 1976.
463. K. Jiraki, Am. J. Forensic Med. Pathol. 13 494. L. Castaner, E. Arrigoni-Martelli, Drugs
(1992) 42–43. Future 2 (1977) 334–337.
464. D. Meyer, V. Halfin, J. Clin. 495. W. F. M. Van Bever, et al.,
Psychopharmacol. 1 (1981) 319–321. Arzneim.-Forsch./Drug Res. 26 (1976)
465. C. F. Seifert, S. Kennedy, Pharmacotherapy 24 1548–1556.
(2004) 776–783. 496. P. G. H. Van Daele et al.,
466. J. G. Stevenson et al., Jt. Comm. J. Qual. Saf. Arzneim.-Forsch./Drug Res. 26 (1976)
30 (2004) 277–281, 233. 1521–1531.
467. A. B. O’Connor, et al., Am. J. Med. 118 (2005) 497. J. E. Leysen et al., Eur. J. Pharmacol. 87
885–889. (1983) 209–225.
468. I.G. Farben, DRP 752 755, 1942. 498. I. P. Monk et al., Drugs 36 (1988) 286–313.
469. H. Kägi, K. Miescher, Helv. Chim. Acta 32 499. M. Goldberg et al., Anesthesiology 63 (1985)
(1949) 2489–2507. 199–201.
94 Analgesics and Antipyretics

500. J. A. Grass, J. Pain Symptom Manage. 7 530. A. A. Lawson, D. B. Northridge, Med. Toxicol.
(1992) 271–286. Adverse Drug Exp. 2 (1987) 430–444.
501. M. Goldberg et al., Anesthesiology 63 (1985) 531. Janssen, US 2 898 340, 1959;
199–201. Searle, US 4 086 234, 1978.
502. Farbw. Hoechst, DBP 865 314, 1941. 532. C. J. Niemegeers et al., Arzneimittelforschung
503. Merck & Co., US 2 644 010, 1953. 22 (1972) 516–518.
504. Abott, US 2 983 757, 1961. 533. D. D. Hung, Anal. Profiles Drug. Subst. 7
505. E. M. Schultz et al., J. Am. Chem. Soc. 69 (1978) 149–169.
(1947) 2454–2459. 534. Janssen, US 3 714 159, 1973;
506. N. R. Easton et al., J. Am. Chem. Soc. 69 ED-OS 2 126 559, 1971.
(1947) 2941–2942. 535. R. A. Stokbroekx et al., J. Med. Chem. 16
507. A. A. Larsen et al., J. Am. Chem. Soc. 70 (1973) 782–786.
(1948) 4195–4196. 536. M. R. Dashwood et al., Prog. Clin. Biol. Res.
508. E. E. Howe, M. Sletzinger, J. Am. Chem. Soc. 328 (1990) 165–169.
71 (1949) 2935. 537. R. C. Heel et al., Drugs 15 (1978) 33–52.
509. M. Bockmühl, G. Ehrhart, Justus Liebigs Ann. 538. R. C. Heel et al., Drugs. 15 (1978) 33–52.
Chem. 561 (1948) 52–85. 539. D. L. DeHaven-Hudkins, et al., Life Sci. 71
510. S. K. Sim, Can. Med. Assoc. J. 109 (1973) (2002) 2787–2796.
615–619. 540. F. Awouters et al., Dig. Dis. Sci. 38 (1993)
511. R. H. Biskara, Anal. Profiles Drug Subs. 3 977–995.
(1974) 365–439. 541. R. C. Heel et al., Drugs. 15 (1978) 33–52.
512. G. D. Olson et al., Clin. Pharmacol. Ther. 21 542. C. D. Ericsson P. C. Johnson, Am. J. Med.
(1977) 147–157. 88(6A) (1990) 10S–14S.
513. M. P.xs Davis, D. Walsh, Support Care Cancer 543. Janssen, DE 1 238 472, 1961.
9 (2001) 73–83. 544. F. Weyne et al., Acta anaesth. belg. 19 (1968)
514. H. Joseph et al., Mt. Sinai J. Med. 67 (2000) 33–45.
347–364. 545. Gödecke, DE 1 518 959, 1965;
515. M. Farell et al., Br. Med. J. 309 (1994) Warner-Lambert, US 3 557 126, 1969.
997–1001. 546. L. E. Overman et al., J. Org. Chem. 44 (1979)
516. M. J. Kreek, JAMA. 223 (1973) 665–668. 4183–4185.
517. J. Lowinson et al., Curr. Psychiatr. Ther. 18 547. G. Satzinger, Justus Liebigs Ann. Chem. 758
(1978) 165–172. (1972) 43–64, 65–71; 728 (1969) 64–87.
518. F. I. Carroll et al., J. Org. Chem. 41 (1950) 548. G. Satzinger et al., Pharm. Ind. 40 (1978)
3521–3524. 657–664.
519. S. A. Walczak et al., Eur. J. Pharmacol. 72 549. R. Schulz et al., Naunyn Schmiedebergs Arch.
(1981) 343–349. Pharmacol. 304 (1978) 89–93.
520. J. D. Blaine et al. Ann. N. Y. Acad. Sci. 362 550. M. Hermann et al., Arzneimittel-Forsch. 20
(1981) 101–115. (1970) 977–983.
521. A. Wolven et al., NIDA Res. Monogr. (1976) 551. K. O. Vollmer, Fortschr. Med. 106 (1988)
29–38. 593–596.
522. J. Kang et al., Eur. J. Pharmacol. 458 (2003) 552. Grünenthal, GB 997 399, 1964;
25–29. US 3 564 100, 1971 (E. Frankus, K. Flick).
523. R. L. Deamer et al., J. Addict Dis. 20 (2001) 553. K. Flick et al., Arzneim.-Forsch./Drug Res. 28
7–14. (1978) no. 1, 107–113.
524. Janssen, DE 1 117 126; 1956; 554. M. C. Frink et al., Arzneimittelforschung 46
GB 822 055, 1956. (1996) 1029–1036.
525. A. J. Grace, Br. Med. J. 281 (1980) 1285. 555. E. Friderichs et al., Arzneimittel-Forsch. 28
526. A. Pohland, H. R. Sullivan, J. Am. Chem. Soc. (1978) 122–134.
75 (1953) 4458–4461; 77 (1955) 3400–3401. 556. R. B. Raffa, E. Friderichs, Pain Rev. 3 (1996)
527. A. Pohland et al., J. Org. Chem. 28 (1963) 249–271.
2483–2484. 557. R. B. Raffa et al., J. Pharmacol. Exp. Ther. 267
528. H. R. Sullivan et al., J. Org. Chem. 28 (1963) (1993) 331–340.
2381–2385. 558. C. R. Lee et al., Drugs 46 (1993) 313–340.
529. D. Boven et al., Hum. Toxicol. 3 (1984) Suppl., 559. K. McClellan, L. J. Scott, Drugs 63 (2003)
1S–238S. 1079–1088.
Analgesics and Antipyretics 95

560. P,Huang et al., J. Pharmacol. Exp. Ther. 297 590. W. K. Schmidt, Drug Alcohol Depend. 14
(2001) 688–695. (1985) 339–362.
561. B. Kögel et al. Eur. J. Pain. 9 (2005) 599–611. 591. G. C. Pugh et al., Br. J. Anaesth. 62 (1989)
562. R. C. Heel et al., Drugs. 17 (1979) 81–110. 601–609.
563. J. Martin, J.Psychoactive Drugs Suppl 2 592. Merck & Co., US 2 364 833, 1944;
(2004) 129–137. US 2 891 954, 1959.
564. S. Sung, J. M. Conry, Ann. Pharmacother. 40 593. M. U. Zubair et al., Anal. Profiles Drug Subs.
(2006) 501–505. 18 (1989) 195–219.
565. T. J. Gal, Clin. Pharmacol. Ther. 45 (1989) 594. Sterling Drug, BE 611 000, 1961.
66–71. 595. S. Archer et al., J. Med. Chem. 7 (1964)
566. N. Griessinger et al., Curr. Med. Res Opin. 21 123–127.
(2005) 1147–1156. 596. R. N. Brogden et al., Drugs 5 (1973) 6–91.
567. A. Dahan et al., Br. J. Anaesth. (2006) 96, 597. D. A. Reed, S. H. Schnoll, JAMA 256 (1986)
627–632. 2562–2564.
598. H. Schmidthammer, Prog. Med. Chem. 35
568. Bristol-Myers, US 3 775 414, 1973.
(1998) 83–132.
569. J. Monkovic et al., J. Am. Chem. Soc. 95
599. G. Ehrhart, H. Ruschig: Arzneimittel,
(1973) 7910.
Entwicklung, Wirkung, Darstellung, vol.1,
570. J. Monkovic et al., J. Am. Chem. Soc. 100
Verlag Chemie, Weinheim 1972, pp. 131–132.
(1978) 4609–4610.
600. J. Hellerbach et al., Helv. Chim. Acta 39
571. C. E. Rosow, Acute Care 12 Suppl 1 (1988)
(1956) 429–440.
2–7. 601. O. Schnider, A. Grüssner, Helv. Chim. Acta 24
572. R. C. Heel et al., Drugs 16 (1978) 473–505. (1951) 2211–2217.
573. B. Ameer, F. J. Salter, Am. J. Hosp. Pharm. 36 602. D. M. Paton, Drugs Future 4 (1979) 438–441.
(1979) 1683–1691. 603. W. Leimgruber et al., Adv. Biochem.
574. R. V. Homan, Am. Fam. Physician 49 (1994) Psychopharmacol. 8 (1973) 45–50.
188–192. 604. F. F. Foldes et al., Can. Anaesth. Soc. J. 16
575. American Home, BE 776 173, 1971; (1969) 151–161.
DE 2 159 324, 1971. 605. J. M. Evans et al., Anaesthesia 29 (1974)
576. M. E. Freed et al., J. Med. Chem. 16 (1973) 721–727.
595–599. 606. US 3 254 088, 1966;
577. J. C. Chen et al., Life Sci. 52 (1993) 389–396. DE 1 183 508, 1962 (M. J. Lewenstein).
578. J. J. O’Brien, et al., Drugs 38 (1989) 226–248. 607. R. A. Olofson et al., Tetrahedron Lett. 1977
579. J. L. Malis, J. Pharmacol. Exp. Ther. 194 1567–1577.
(1975) 488–498. 608. R. Simantov, S. H. Snyder, Biochem. Soc.
580. J. J. O’Brien, P. Benfield, Drugs 38 (1989) Trans. 5 (1977) 62–65.
226–248. 609. K. A. Handal et al., Ann. Emerg. Med. 12
581. Wyeth, DE-OS 1 941 534, 1969; (1983) 438–445.
GB 1 285 025, 1969. 610. P. M. Goodrich, AANA J. 58 (1990) 14–16.
582. G. Bradley et al., Eur. J. Med. Chem. 15 (1980) 611. Endo Laboratories, GB 1 119 270, 1968;
375. US 3 332 950, 1967.
583. B. Holmes, A. Ward, Drugs 30 (1985) 612. V. Höllt, A. Herz, Fed. Proc. 37 (1978)
285–312. 158–161.
584. G. W. Pasternak et al., Postgrad. Med. J. 61 613. B. L. Crabtree, Clin. Pharm. 3 (1984)
Suppl 2 (1985) 5–12. 273–280.
585. B. Holmes, A. Ward, Drugs 30 (1985) 614. J. P. Gonzalez, R. N. Brogden, Drugs 35
285–312. (1988) 192–213.
586. Endo Laboratories, GB 1 119 270, 1968; 615. B. A. Judson et al., Drug Alcohol Depend. 7
US 3 332 950, 1967. (1981) 325–346.
616. Boehringer Ingelheim Pharma GmbH & Co.
587. L. Castaner, P. J. Roberts, Drugs Future 2
KG, US 4176186, 1979 (L. Goldberg, H.
(1977) 613–615.
Merz, K. Stockhans).
588. E. B. De Souza, et al., J. Pharmacol. Exp.
617. C. S. Yuan, J. F. Foss, Neuropharmacology 38
Ther. 244 (1988) 391–402.
(1999) 425–432.
589. J. K. Errick, R. C. Heel, Drugs 26 (1983)
618. C. S. Yuan et al., J. Pharmacol. Exp. Ther. 300
191–211.
(2002) 118–123.
96 Analgesics and Antipyretics

619. D. S. Wang et al., J. Emerg. Med. 16 650. H. A. Friedel, A. Fitton, Drugs 45 (1993)
(1998):471–475. 548–569.
620. H. E. Jones et al., Drug Alcohol Depend. 60 651. Pfizer, US 4152326, (J. Hartenstein, G.
(2000) 29–37. Satziger).
621. L. C. Costantini et al., Int. J. Pharm. 283 652. Pfizer, US 4024175, (G. Satzinger, J.
(2004) 35–44. Hartenstein, M. Hermann, W. Heldt).
622. J. E. Grant et al., Am. J. Psychiatry 163 (2006) 653. Zambon Group, WO 2005 044 779, 2005 (K.
303–312. Arrighi, F. Corcella, G. Marchioro, A. Nicoli,
623. K. Hempenstall, A. S. Rice, Curr. Opin. M. Paiocchi, M. Villa).
Investig. Drugs 3 (220) 441–448. 654. Taro Pharmaceutical Industries Ltd., WO 2003
624. R. H. Dworkin et al., Arch. Neurol. 60 (2003) 089403, 2003
1524–1534. 655. R. Cagnoli et al., Tetrahedron 59 (2003) 9951.
625. H. Ueda, Pharmacol Therap. 109 (2006) 656. G. J. Sills, Curr. Opin. Pharmacol. 6 (2006)
57–77. 108–113.
626. D. B. Gordon, G. Love, Pain Manage. Nurs. 5 657. J. K. Baille, I. Power Curr. Opin. Investig.
(2004) 19–33. Drugs 7 (2006) 33–39.
627. G. T. Carter, M. D. Sullivan, Curr. Opin. 658. I. W. Tremont-Lukats et al., Drugs 60 (2000)
Investig. Drugs 3 (2002) 454–458. 1029–1052.
628. J. Zhou, Drugs of the Future 29 (2004) 659. J. Devulder et al., Acta Neurol. Belg. 102
1235–1244. (2002) 97–103.
629. J. Lai et al., Ann. Rev. Pharmacol. Toxicol. 44 660. Wellcome Foundation, EP 21 12, 1981 (M. G.
(2004) 371–397. Baxter, et al.).
630. Ciba, US 3 471 548. 1969. 661. Wellcome, US 4 560 687, 1985 (M. G. Baxter,
631. Ciba, US 3 978 216, 1972. A. R. Elphick, A. A. Miller, D. A. Sawyer).
632. N. G. Bowery, Curr. Opin. Pharmacol. 6 662. Wellcome, US 4 602 017, 1986 (M. G. Baxter,
(2006) 37–43. A. R. Elphick, A. A. Miller, D. A. Sawyer).
633. Geigi, US 2 948 718, 1960 (W. Schindler). 663. E. Eisenberg et al., Rev. Neurotherapeutics 5
634. DD 133 052, 1977 (R. Müller). (2005) 729–735.
635. D. Soyka et al., Schmerz 12 (1998) 419–427. 664. M. Guven, H. Bozdemir, J. Gunay, Y. Sarica, J.
636. Boehringer Ingelheim, DE 1 303 141, 1961; Kahraman, F. Koc, The actions of lamotrigine
US 3 236 857, 1966. and levetiracetam on the conduction properties
637. VEB Arzneimittelwerke Dresden, DE-AS 1 of isolated rat sciatic nerve, Eur. J. Pharmacol.
770 874, 1968. 553 (2006), 129–134.
638. H. Schmitt, Handb. Exp. Pharmacol. 39 665. Rexall, US 3 487 153, 1969.
(1977) 299–396. 666. Riker, US 3 830 803, 1974.
639. C. Lund et al., Br. J. Anaesth. 63 (1989) 667. R. C. Heel et al., Drugs 19 (1980) 249–267.
516–519. 668. Park Davis, US 2 409 754, 1946 (H. R. Henze).
640. D. R. Jasinki et al., Arch. Gen. Psychiatry 42 669. H. Biltz et al., Ber. Dtsch. Chem. Ges. 41
(1985) 1063–1066. (1908) 1391.
641. J. Deeter et al., Tetrahedron Lett. 31 (1990) 670. Warner-Lambert WO 99/48876, 1999 (R. L.
7101. Bosch)
642. L. A. Sorbera et al., Drugs of the Future 25 671. B. D. Maryanoff, B. L. Margul, Drugs of the
(2000) 907–916. Future 14 (1989) 342.
643. H. Liu et al., Chirality 12 (2000) 26–29. 672. B. E. Maryanoff, et al., Med. Chem. 30 (1987)
644. S. Attila, E. Leinonen, Curr. Opin. 880–887.
Investig.Drugs 3 (2002) 1217–1221. 673. Ortho-McNeil Pharmaceuticals Inc., US
645. N. Pitsikas, Curr. Opin. Investig. Drugs 1 4513006, 1985 (B. E. Maryanoff, J. F.
(2000) 116–121. Gardocki).
646. Degussa, DE 1 795 858, 1968; 674. Ortho-McNeil Pharmaceuticals Inc., WO 2004
US 3 513 171, 1970. 078769, 2004 (J. E. Berkner, S. Duncan)
647. Drugs Future 8 (1983) 773–775. 675. C. E. Heading, IDrugs 4 (2001) 339–350.
648. W. von Bebenburg et al., Chem. Ztg. 103 676. C. E. Heading, Curr. Opin. Investig. Drugs 3
(1979) 387–399. (2002) 915–920.
649. W. von Bebenburg et al., Chem. Ztg. 105 677. S. Tries et al., Inflamm. Res. 51 (2002)
(1981) 217–219. 129–134.
Analgesics and Antipyretics 97

678. J. J.Talley, .J Med. Chem. 43 (2000) 775–777. 681. Troponwerke, US 3 692 818, 1972,
679. US 2 806 033, 1955 (M. J. Lewenstein, U. DE 1 939 112, 1969.
Weiss). 682. Syntex, US 3 896 157, 1975.
680. T. Appelboom, P. Franchimont, Adv. Therapy 683. Geigi, US 3 558 690, 1971.
11 (1994) 228–234.

You might also like