22.05.2022 Views

DƯỢC LÍ Goodman & Gilman's The Pharmacological Basis of Therapeutics 12th, 2010

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

The postsynaptic category includes all the events that follow

release of the transmitter in the vicinity of the postsynaptic receptor.

Examples include the molecular mechanisms by which receptor

occupancy alters the properties of the membrane of the postsynaptic

cell (shifts in membrane potential), as well as more enduring biochemical

actions (e.g., changes in second messenger concentrations,

protein kinase and phosphoprotein phosphatase activities, and phosphoprotein

formation). Direct postsynaptic effects of drugs generally

require relatively high affinity for the receptors or resistance to

metabolic degradation. Each of these presynaptic or postsynaptic

actions is potentially highly specific and can be envisioned as

restricted to a single, chemically defined subset of CNS cells.

Convergence, Synergism, and Antagonism Result from

Transmitter Interactions. Although the power of the reductionist

approach to clone cDNAs for receptors or receptor subunits and to

determine their properties by expression in cells that do not normally

express the receptor or subunit cannot be underestimated, the simplicity

of cell culture models may not reproduce the nuances of

receptor function in vivo and may divert attention from the complexity

of the intact CNS. A given neurotransmitter may interact simultaneously

with all of the various isoforms of its receptor on neurons

that also are under the influence of multiple other afferent pathways

and their transmitters. Thus, the use of model systems to predict the

behavioral or therapeutic consequences of drugs in humans may fail

as a consequence of the complexity of the interactions possible,

including differences between normal and diseased tissue.

CNS Drug Discovery. As is evident from the chapters to follow, a

large number of agents have been developed to treat neuropsychiatric

diseases. With few exceptions these agents offer primarily

symptomatic improvement; few are truly disease modifying. For

example, the use of L- dopa to treat Parkinson disease alleviates the

symptoms effectively but the disease continues to progress.

Similarly, although antipsychotics and antidepressants are often efficacious,

the symptoms tend to recur. Moreover, many drugs developed

to treat CNS diseases are not uniformly effective:

Approximately one- third of patients with severe depression are

“treatment-resistant.” CNS diseases are complex, with multiple

symptoms, some of which, like the negative symptoms of schizophrenia,

tend to be resistant to treatment. Furthermore, the complexity

of the brain and its neuronal pathways results in significant risk

of side effects, even when the most biochemically selective agent is

administered. Particularly disquieting side effects include tardive

dyskinesias that can result from prolonged treatment with antipsychotics

and the hypnotic effects of agents used for the treatment of

seizure disorders.

The lack of disease- modifying treatments for CNS diseases

represents a very significant unmet medical need and makes the discovery

of such agents a high priority. Drug discovery is generally a

high- risk/high- reward effort (Chapter 1) and CNS- active agents are

no exception. Overall, the probability of success in developing a drug

from the time a compound enters into clinical trials is ~10%; the

success rate for CNS drugs is somewhat lower. Myriad factors contribute

to the increased difficulty and reduced probability of success

in efforts to develop drugs to treat CNS disease. Factors that reduce

the probability of success include the complexity of neural pathways

governing behavior and its pathology, and the permeability barriers

that restrict access of drugs to CNS sites (including the blood- brain

barrier and drug export systems; see Chapters 2 and 5). For example,

a drug that affects serotinergic transmission may affect 14 5-HT

receptor subtypes that are involved in a plethora of biological systems.

Moreover, animal models of CNS diseases are often incompletely

validated and an effect in an animal model may not be

predictive of efficacy in human disease. For instance, a number of

agents reduce the size of the infarct when given to animals following

occlusion of the middle cerebral artery, yet none of these agents

has shown positive results in human clinical trials. A number of

agents that inhibit the reuptake of 5-HT and NE are effective in treating

depression. In a number of animal models of depression, these

agents produce an effect after a single dose, which corresponds

nicely with the time course for the inhibition of neurotransmitter

reuptake in experimental animals, whereas, a period of several weeks

is typically required to see a therapeutic effect in humans. Given this

discrepancy, one can understand the reluctance of pharmaceutical

companies to invest in clinical trials of new antidepressants on the

basis of effects in animal models.

Clinical trials represent another area of challenge in developing

new therapies for CNS diseases. For example, testing an agent to

treat depression likely requires a trial lasting 6 or more weeks and the

placebo response rate may exceed 50%. Such conditions necessitate

large, prolonged, and therefore expensive trials. Studies of treatments

for neurodegenerative disease are even more difficult. With current

diagnostic capabilities, it is difficult to detect a significant change in

the rate of progression of cognitive decline in patients with

Alzheimer’s disease in less than a year. One way to circumvent the

long time necessary to detect a biological meaningful result is

through the use of surrogate markers (e.g., a decrease in serum cholesterol

for improved cardiovascular morbidity and mortality).

Regrettably, there are relatively few useful surrogate markers for

CNS diseases.

Impact of Genomics on CNS Drug Discovery. The sequencing of

the human genome has the potential to significantly change drug

discovery in the CNS. Thus, genetic testing may predict the likelihood

that a given individual will develop a particular disease, will

respond to a particular therapy, or will suffer side effects from a

particular treatment paradigm. Genetic testing may be particularly

important in the case of CNS diseases, where the etiology is likely

to be multigenic. Molecular approaches will likely speed development

of more and improved animal models that better mimic

human disease.

BIBLIOGRAPHY

Aimone JB, Deng W, Gage FH. Adult neurogenesis: integrating

theories and functions. Trends Cogn Sci, 2010, 14: in press.

Alexander SPH, Mathie A, Peters JA. Guide to Receptors and

Channels. Br J Pharmacol, 2006, 147(s3):S1.

Aston- Jones G, Chen S, Zhu Y, Oshinsky ML. A neural circuit

for circadian regulation of arousal. Nat Neurosci, 2001,

4:732–738.

Bettler B, Kaupmann K, Mosbacher J, Gassmann M. Molecular

structure and physiological functions of GAGA B

receptors.

Physiol Rev, 2004, 84:835–867.

Biel M, Michalakis S. Cyclic nucleotide-gated channels.

Handb Exp Pharmacol, 2009, 191: 111–136.

393

CHAPTER 14

NEUROTRANSMISSION AND THE CENTRAL NERVOUS SYSTEM

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!