Sunteți pe pagina 1din 61

Pengembangan Obat

FA-5002 (3 sks)

Aspek Umum & Regulasi

Dosen: Tutus Gusdinar Kartawinata


Laboratorium Biokimia Medik & Kimia Klinik
Kelompok Keilmuan Farmakokimia
Sekolah Farmasi – Institut Teknologi Bandung
gusdinar@fa.itb.ac.id
NO TOPIK DOSEN
01 Aspek Umum dan Regulasi terkait dengan Pengembangan Obat TG

02 Penemuan Obat Baru & Ilmu Penunjang Riset TG

03 Konsep Biokimia Reseptor (Reseptorologi) TG

04 Konsep Biokimia Protein Farmaka (Struktur 3D & Teknik Pemurnian) TG

05 Desain dan Penemuan Obat untuk Negara Berkembang TG

06 Kajian Keamanan Obat IK

07 Pedoman Uji Klinik Obat yang Baik IK

08 Pengembangan Obat Baru IK

09 Sekilas Bahan Alam SX

10 Ekstrak Bahan Alam & Komatografi Ekstrak SX

11 High Through put Screening as a Rout to Discovering SX

12 Dasar Pengembangan Formulasi Obat Baru STD/DM

13 Desain dan Evaluasi Produk Obat STD/DM

14 Biofarmasetika DSR
Legislasi dan regulasi obat merupakan
bagian dari sistem pelayanan kesehatan

• “Drugs are a public good and not simply just another


commodity: first for their high social value, and then
because consumers and prescribers are unable to
assess their quality, safety and efficacy.”
Dr. Gro Harlem Brundtland
Director General of WHO EDM
Drug discovery and preclinical trials

The development of new drugs over the past


30 years has revolutionalized the practice of
medicine and has for instance seen the
increased use of new antihypertensives and
drugs that reduce cholesterol synthesis or
dissolve blood clots which led to a 50%
reduction in the number of deaths from
cardio-vascular diseases and stroke among
other diseases.
Conventional approaches to Drug discovery
1. Traditional knowledge approach (pendekatan pengetahuan tradisi)
This is the discovery of drugs based on traditional medical knowledge.
The best example is the documented analgesic effects of extracts from
opium poppy that led to the isolation of morphine from the plant and
the subsequent synthesis of related analgesics.
2. Discovery through serendipity (penemuan secara tidak disengaja)
This is the accidental discovery of novel drugs based on the ingenuity of
a scientist investigating a problem initially unrelated to the observed
phenomenon; examples of such discoveries include the observation by
Alexander Flemmings that penicillium mould could inhibit the growth of
bacteria. This finding led to the discovery of antibiotics. Discovery of
therapeutic usefulness of a side effect e.g. clonidine originally used as a
nasal decongestant was found to have antihypertensive properties
while, the hypoglycemic effects of sulphonamides used in the treatment
of typhoid fever led to the development of structurally related
sulphonylureas as oral hypoglycemic drugs.
3. Discovery from effects of endogenous agents in test animals
An example of discovery arising from studies of endogenous
agents in test animals is the anticoagulant action of the
venom from the Malayan viper that led to the identification
of the anticoagulant ancrod.
4. Modern approaches to drug discovery
These are those approaches that form a basis for the rational
design of drugs.
5. Bioprospecting
This is the screening of a large number of natural products,
chemical entities, large libraries of peptides, nucleic acids
and other organic molecules for biological activity. This
approach may lead to identification and development of
new drug molecules.
6. Metabolomics
This is the profiling of natural products of related plant
species screening using either liquid or gas chromatography
mass spectrometry to determine active metabolites that
may be present in novel crude herbal medical preparations.
7. In silico screening (penapisan dengan piranti-lunak
komputer)
This is the most advanced technique for drug discovery. It
entails virtual screening or docking of compounds on the 3-
D- structure of a known receptor based on homologies of
the test drug molecules with a known test parent drug. In
silico screening can form a basis for the modification of a
known drug molecule to determine possible therapeutic
applications and may lead to the development of putative
drugs against new targets.
The use of the suffix ‘ome’ is defined in the Oxford English
dictionary as being used in cellular and molecular biology to
form nouns with the sense ‘all constituents considered
collectively’. Interestingly, the use of the ‘ome’ suffix in biology
dates back to the early 1900s when it was first used to describe a
‘biome’ and genome (although originally in German as genom).

Over the past few decades, the use of the omics suffix has rapidly
increased, due in a large part, to the rapid growth in technologies
that allow global analysis of samples on a systems level.

The familiarity of the ‘omics’ term also was rapidly advanced by


the completion of the human genome in the early 2000s. Since
that time, we have seen the term ‘omics’ move beyond use for
genomics and proteomics and gain acceptance for a diverse array
of systems biology applications.
8. Screening of putative drug molecules (putative=diduga)
Selection of molecules for further study is usually conducted in animal models
of human disease and the pharmacological tests include both the in vitro
and in vivo studies after the initial screening for biological activity. For
instance, antibacterial activity of drugs is assessed by their ability to inhibit
growth of a variety of microorganisms, while hypoglycemic drugs are tested
for their ability to lower blood pressure.
The in vitro methods include incubation of a parent compound with various
subcellular fractions such as microsomes, individual recombinant drug
metabolizing enzymes from cells or tissue slices. The in vivo studies involve
working on typical animal models such as dogs or rats. Some of the in vitro
and in vivo studies that may be performed

If an agent possesses useful activity it would be further studied for


possible adverse effects on other major organs. These studies might
suggest the need for further chemical modification to achieve desirable
pharmacokinetic/ pharmacodynamic properties (pharmacomodulation).
The R &D process Development

Preclinical studies Clinical studies


Discovery Early Clinical Development
CHEMISTRY/ IND* PHASE I PHASE II PHASE III NDA** PHASE IV
PHARMA-
COLOGY
Search for Regulatory Efficacy Clinical Comparative Regulatory Continued
active review studies on studies on a studies on a review comparative
substances *Investigational healthy limited scale large studies
New Drug volunteers number of
Toxicology, KNOWLEDGE
Application for
50–150 100–200 patients
efficacy permission to LEVEL Registration,
studies on administer a new persons patients
market
various types drug to humans 500–5,000
introduction
of animals KNOWLEDGE patients
**New Drug
LEVEL Application
Application for
permission to market
a new drug

TIME SPAN

2–4 yrs. 2–6 months 3–6 yrs. 1–3 yrs.

Approximately 10–15 years from idea to marketable drug

13/09/2017 16
Active Pharmaceutical Ingredient (API)
Development, Characterization
API process development Discovery route assessment
• Route scouting
• Lab scale API batch manufacture
• IND route development
• Primary reference synthesis & characterizations
• Preparation of major impurities in API and characterizations
• NDA route development
• Process safety studies
• Process development to support multi-kilo manufacture with
optimization
• API preformulation
• Crystallization process development leading to the desired API
physico-chemical properties.
API synthesis and supply

API analytical development and support

API characterization Appearance


• Elemental analysis
• Spectroscopic methods (IR, UV-VIS, NMR, Mass Spec)
• Purity (HPLC, GC, LC/MS/MS, etc.)
• Chirality (optical rotation)
• Residual solvents & water content
• Thermogravimetry, DSC (Endothermic/ Exothermic properties)
• SEM (scanning electron microscope)
• Optical microscopy
• Particle size distribution
• Crystalline vs. amorphous by X-ray powder diffraction (XRPD)
• Polymorphism by X-ray powder diffraction (XRPD)
• Stability (susceptibility for oxidation, hydrolysis, photochemical
reactions, etc.)
Formulation and Preformulation
Development
• Preformulation
– Salt screening
– Polymorph screening
– Solubility
– API – excipient interaction
– Novel technologies for highly insoluble APIs (nano technology)
• Formulation development
– IV and oral preclinical formulations
– Clinical formulations
• Solid
• Semi-solid
• Oral Liquids
• Sterile (aseptically prepared) liquids
– Process development studies
– 14C labeled and cytotoxic formulations
– Process scale-up studies
• Analytical support for preformulation and formulation development, including
method development and validation
• Microbiological testing
Key facilities and instrumentation
Highlights of key facilities and instrumentation:
• Separate laboratories for non-radiolabelled and radiolabeled materials
• X-Ray powder diffraction system
• Thermo-gravimetric analysis, differential scanning calorimetry
• 400 MHz and 500 MHz NMR spectrometers
• Solid state NMR
• ICP-MS / LC-ICP-MS
• Variety of spectrometers and mass spectrometers
• Variety of chromatography systems with variety of detection systems
• Hot stage microscope
• Dynamic vapor absorption/desorption system
• ICH compliant stability suite
CMC Regulatory Documentation
(Chemistry, Manufacturing and Controls processes)
• Preparation of CMC documents for inclusion in regulatory dossiers, including
coordination, writing and compilation
– Global marketing applications using the e-CTD format
– Investigational Medicinal Product Dossiers (IMPD)
– Investigational New Drug Applications (IND)
– Quality Overall Summaries (QOS)
– Clinical Investigator Brochures (CIBs)
– Integrated drug development reports
– Release testing monographs for APIs and drug products
– Interaction with other disciplines such as toxicology and drug metabolism to ensure
consistency of dossier sections
• Review of technical CMC documents either authored in house or as a service
to increase your probability of success with the regulatory agencies
• Consultation and response to questions from agencies
– Responding to regulatory CMC questions from global health authorities
– Preparation of position papers and responses for potential regulatory questions
• Production of IMPDs or INDs for the clinical administration of radiolabeled
species
DRUG DEVELOPMENT
Drug development is a blanket term used to
define the entire process of bringing a new drug
or device to the Market. It includes drug
discovery/product development, pre-clinical
research(microorganisms/animals) and clinical
trials (on humans).
Few people still refer to the drug development as
mere preclinical development.
NCE DEVELOPMENT
• New chemical entities (NCEs) are compounds which
emerge from the process of drug discovery. These will
have promising activity against a particular biological target
thought to be important in disease; however, little will be
known about the safety, toxicity, pharmacokinetics and
metabolism of this NCE in humans.

• It is the function of drug development to assess all of


these parameters prior to human clinical trials. A further
major objective of drug development is to make a
recommendation of the dose and schedule to be used the
first time an NCE is used in a human clinical trial "first-in-
man" (FIM) or First Human Dose (FHD).
NCE DEVELOPMENT

• In addition, drug development is required to establish the


physicochemical properties of the NCE: its chemical
makeup, stability, solubility. The process by which the
chemical is made will be optimized so that from being
made at the bench on a milligram scale by a synthetic
chemist, it can be manufactured on the kilogram and then
on the ton scale. It will be further examined for its
suitability to be made into capsules, tablets, aeresol,
intramuscular injectable, subcuteneous injectable, or
intravenous formulations. Together these processes are
known in preclinical development as CMC (Chemistry,
Manufacturing and Control).
NCE DEVELOPMENT

• Many aspects of drug development are focused on


satisfying the regulatory requirements of drug
licensing authorities. These generally constitute a
number of tests designed to determine the major
toxicities of a novel compound prior to first use in
man.

• It is a legal requirement that an assessment of major


organ toxicity be performed (effects on the heart and
lungs, brain, kidney, liver and digestive system), as
well as effects on other parts of the body that might
be affected by the drug (e.g. the skin if the new drug
is to be delivered through the skin).
NCE DEVELOPMENT

• While, increasingly, these tests can be made


using in vitro methods (e.g. with isolated cells),
many tests can only be made by using
experimental animals, since it is only in an
intact organism that the complex interplay of
metabolism and drug exposure on toxicity can
be examined.
NCE DEVELOPMENT

• The process of drug development does not stop


once an NCE begins human clinical trials. In
addition to the tests required to move a novel
drug into the clinic for the first time it is also
important to ensure that long-term or chronic
toxicities are determined, as well as effects on
systems not previously monitored (fertility,
reproduction, immune system, etc). The
compound will also be tested for its ability to
cause cancer (carcinogenicity testing).
NCE DEVELOPMENT

• If a compound emerges from these tests with an


acceptable toxicity and safety profile, and it can
further be demonstrated to have the desired
effect in clinical trials, then it can be submitted
for marketing approval in the various countries
where it will be sold. In the US, this process is
called a New Drug Application or NDA.
• Most NCEs, however, fail during drug
development, either because they have some
unacceptable toxicity, or because they simply do
not work in clinical trials.
NCE DEVELOPMENT

• As this drug discovery process becomes more expensive it


is becoming important to look at new ways to bring
forward NCEs. One approach to improve efficiency is to
recognize that there are many steps requiring different
levels of experimentation. The early phase of drug
discovery actually has components of real innovation,
components of experimentation and components that
involve set routines.
• This model of Innovation, Experimentation, and
Commoditization ensures that new ways to do work are
adopted continually. This model also allows the discipline
to use appropriate internal and external resources for the
right work.
COST ?
• Studies published in 2003 report an average pre-tax cost of
approximately $800 million to bring a new drug (i.e. a drug with
a New Chemical Entity) to market.
• A study published in 2006 estimates that costs vary from around
500 million to 2,000 million dollars depending on the therapy
or the developing firm.
• These figures relate only to new, innovative drugs (drugs with a
New Chemical Entity NCE, also called New Active Substance
NAS). Each year, worldwide, only about 26 such drugs enter the
market (2005: 26, 2004: 24, 2003: 26, 2002: 28). The
development cost of the thousands of other drugs are much
smaller. The $800 million quoted include the cost of all drug
development which did not result in a new drug. It also includes
some $400 million of opportunity costs.
CLINICAL TRIAL
• Clinical trials are conducted to allow safety and
efficacy data to be collected for new drugs or
devices. These trials can only take place once
satisfactory information has been gathered on the
quality of the product and its non-clinical safety,
and Health Authority/Ethics Committee approval
is granted in the country where the trial is taking
place.
CLINICAL TRIAL

• Depending on the type of product and the stage


of its development, investigators enroll healthy
volunteers and/or patients into small pilot studies
initially, followed by larger scale studies in
patients that often compare the new product with
the currently prescribed treatment. As positive
safety and efficacy data are gathered, the number
of patients is typically increased.
• Clinical trials can vary in size from a single center
in one country to multicenter trials in multiple
countries.
CLINICAL TRIAL

• Due to the sizable cost a full series of clinical


trials may incur, the burden of paying for all the
necessary people and services is usually borne
by the sponsor who may be a governmental
organization, a pharmaceutical or biotechnology
company.
• Since the diversity of roles may exceed
resources of the sponsor, often a clinical trial is
managed by an outsourced partner such as a
contract research organization.
Drugs of the future
Small-chemical drugs will be te principal
pharmaceutical tools for the foreseeable
future, though with monoclonal antibodies
and proteins making an increasing impact.
Gene therapy
• In the 1980s, there were high hopes that gene therapy would
open up a wealth of new treatments, particularly for inherited
conditions. The idea is that a gene is delivered into cells and
begins to make a therapeutic protein. So people with cystic
fibrosis, who lack a working version of a protein known as
CFTR, would receive a copy of the CFTR gene.
• Unfortunately, the promise has yet to be realised. It has proved
difficult to get active DNA into the nucleus and stably active.
Progress has been slower than expected, and also suffered after
the death of a patient, Jesse Gelsinger, in a clinical trial in 1999.
A further setback came in 2003, when French patients developed
cancer linked to the integration of a viral vector into their DNA.
• Nevertheless, clinical trials are underway in a number of
conditions, including muscular dystrophy and Parkinson's
disease. Gene therapy is also being tested in some cancers,
though the aim is to kill cells rather than repair them. Routine
use, however, remains a long way off.
RNAi
• RNA interference (RNAi), which gained a Nobel Prize for its
discoverers in 2006, is a new and highly promising strategy. RNAi
is used to eliminate (or 'knock down') specific proteins from a
cell, such as those causing a disease. It is based on an unusual
phenomenon: short RNA molecules triggering highly specific
destruction of messenger RNA molecules containing the same
RNA sequence. Its normal role is probably to protect against
viruses invading the cell.
• The medical possibilities are very broad. Examples include
knocking down the receptor for a virus, or an overactive protein
causing cancer or messenger molecules promoting inflammation.
• A small number of clinical trials have begun, for example for
macular degeneration (a form of blindness). But it is early days.
As in gene therapy, it is difficult to deliver the RNA and there are
worries that other, useful proteins might be eliminated. One
study in mice led to severe liver damage in animals, possibly
because large doses of RNA were used.
Nanotechnology
• Nanotechnology-based solutions are being tested in a variety
of conditions.
• Some applications depend on the unusual properties of
materials at the nanoscale. Nanoscale silver is toxic to bacteria
and is being used in wound dressings (silver-impregnated
pyjamas have been suggested for hospitals). Gold
nanoparticles can convert some wavelengths of light into
intense heat, and are being tested as a possible cancer
treatment (a 'thermal scalpel').
• Critical to many applications will be targeting. Antibodies
could target a toxin-linked nanoparticle to a cancer cell.
• More generally, because they are so small, weight-for-weight
nanoparticles have a very high surface area. There is interest
in using this property for controlled release of drugs.
• Nanobased structures are being explored as molecular scaffolds for tissue
repair. Some exciting applications combine a physical support role for
nanomaterials with bioactive molecules attached to a nanoscale scaffold. This
approach could be used to encourage bone or nerve growth following tissue
damage.

• Nanotechnologies also show significant promise in diagnostics (for example,


through 'lab-on-a-chip' technologies, or by detection of very low
concentrations of key metabolites) and medical imaging. Another exciting
possibility is to link detection to treatment - so a diagnostic device
automatically delivers the required medication. In animal studies,
nanoparticles have been used both to detect blood glucose levels and to
release insulin.

• Nanotechnologies are undoubtedly an area of great promise. Given the


diversity of approaches they encompass, they could have a profound impact
on healthcare. Initially they may enhance current treatments, but entirely new
agents could soon become available.

• However, nanotechnologies also raise challenging regulatory issues - the


properties of nanomaterials differ fundamentally from their everyday
counterparts; can they be considered the same substance? And there are
concerns about the possible environmental impact of nanoparticles.
A living thing
As well as chemically produced agents, researchers are also looking at living organisms.
In doing so, they are reviving a long and colourful medical history.
• Leeches may not be everyone's cup of tea, but they produce a very useful anti-blood-
clotting agent (hirudin) and are very effective at draining blood. They are used clinically in
microsurgery, helping to improve blood flow when digits are reattached.
• Maggots may be similarly repellent to most, but they have long been medicinally useful. In
World War I, infections with maggots kept bacterial infections in check. Experiments have
been carried out with maggots to clean wounds; they also seem to secrete compounds that
promote wound healing. They have been shown to be just as good (and cost-effective) as
conventional medications for chronic wounds, and greenbottle larvae are commercially
available for use in medicine. The main obstacle to their wider use is patient
squeamishness…
• An area of growing interest is the use of parasites or their secretions or eggs to manipulate
the immune response. There is a school of thought that the current high incidence of
asthma, inflammation and allergy in the West is due to a lower parasite burden. In parts of
the world where parasites are common, asthma is rare. Various trials have been carried out
of parasitic worm eggs for inflammatory bowel disease, with some success. In the UK,
hookworms are being tested as a treatment for asthma.
• Much effort is being put into identifying the active substances produced by parasites, so
that they can be given medicinally without a patient having to be infected with the real thing
Teori Dasar untuk Memahami Filosofi
Riset dan Pengembangan Obat
[Drug R&D]

I. Cell Communication and Cell Signaling


II. Molecular Drug – Receptor Interaction
III.Biocatalytic Mechanism of Enzyme
CELL COMMUNICATION
&
CELL SIGNALING
CELL SIGNALING
• Cell signaling is part of a complex system of
communication that governs basic cellular activities
and coordinates cell actions. The ability of cells to
perceive and correctly respond to their micro-
environment is the basis of development, tissue repair,
and immunity as well as normal tissue homeostasis.
Errors in cellular information processing are
responsible for diseases such as cancer, autoimmunity,
and diabetes.
• By understanding cell signaling, diseases may be
treated effectively and, theoretically, artificial tissues
may be yielded.
• Traditional work in biology has focused on studying
individual parts of cell signaling pathways. Systems
biology research helps us to understand the underlying
structure of cell signaling networks and how changes in
these networks may affect the transmission and flow of
information. Such networks are complex systems in
their organization and may exhibit a number of
emergent properties including bistability and
ultrasensitivity. Analysis of cell signaling networks
requires a combination of experimental and
theoretical approaches including the development and
analysis of simulations and modelling.
RECEPTORS FOR CELL SIGNAL
• Cells receive information from their environment
through a class of proteins known as receptors. Notch is a
cell surface protein that functions as a receptor. Animals
have a small set of genes that code for signaling proteins
that interact specifically with Notch receptors and
stimulate a response in cells that express Notch on their
surface. Molecules that activate (or, in some cases,
inhibit) receptors can be classified as hormones,
neurotransmitters, cytokines, growth factors but all of
these are called receptor ligands. The details of ligand-
receptor interactions are fundamental to cell signaling.
RECEPTORS FOR CELL SIGNAL

• While many receptors are cell surface proteins,


some are found inside cells. For example,
estrogen is a hydrophobic molecule that can
pass through the lipid bilayer of cell surface
membranes. Estrogen receptors inside cells of
the uterus can be activated by estrogen that
comes from the ovaries, enters the target cells,
and binds to estrogen receptors.
RECEPTORS FOR CELL SIGNAL

• A number of transmembrane receptors for


molecules that include peptide hormones and of
intracellular receptors for steroid hormones exist,
giving to a cell the ability to respond to a great
number of hormonal and pharmacological
stimuli. In diseases, often, proteins that interact
with receptors are aberrantly activated, resulting
in constitutively activated downstream signals.
RECEPTORS FOR CELL SIGNAL

• For several types of intercellular signaling


molecules that are unable to permeate the
hydrophobic cell membrane due to their
hydrophilic nature, the target receptor is
expressed on the membrane. When such
signaling molecule activates its receptor, the
signal is carried into the cell usually by means
of a second messenger such as cAMP.
CELLS COMMUNICATION
Within endocrinology (the study of intercellular signalling in animals) and the
endocrine system, intercellular signalling is subdivided into the following
classifications:

• Endocrine signals are produced by endocrine cells and travel


through the blood to reach all parts of the body.
• Paracrine signals target only cells in the vicinity of the emitting
cell. Neurotransmitters represent an example.
• Autocrine signals affect only cells that are of the same cell type
as the emitting cell. An example for autocrine signals is found in
immune cells.
• Juxtacrine signals are transmitted along cell membranes via
protein or lipid components integral to the membrane and are
capable of affecting either the emitting cell or cells immediately
adjacent.
Many cell signals are carried by molecules that are released by one
cell and move to make contact with another cell.
Endocrine signals are called hormones. Hormones are produced
by endocrine cells and they travel through the blood to reach all
parts of the body. Specificity of signaling can be controlled if only
some cells can respond to a particular hormone.
Paracrine signals target only cells in the vicinity of the emitting cell.
Neurotransmitters represent an example. Some signaling
molecules can function as both a hormone and a neurotransmitter.
For example, epinephrine and norepinephrine can function as
hormones when released from the adrenal gland and are
transported to the heart by way of the blood stream.
Norepinephrine can also be produced by neurons to function as a
neurotransmitter within the brain. Estrogen can be released by the
ovary and function as a hormone or act locally via paracrine or
autocrine signaling.
Juxtacrine signaling is a type of intercellular communication that is
transmitted via oligosaccharide lipid, or protein components of
a cell membrane, and may affect either the emitting cell or the
immediately-adjacent cells.
It occurs between adjacent cells that possess broad patches of
closely-opposed plasma membrane linked by transmembrane
channels known as connexons. The gap between the cells can
usually be between only 2 – 4 nm.
Unlike other types of cell signaling (such as paracrine and
endocrine), juxtacrine signaling requires physical contact
between the two cells involved.
Juxtacrine signaling has been observed for some growth factors,
cytokine and chemokine cellular signals.
Autocrine signaling is a form of signalling in which a cell
secretes a hormone or chemical messenger (called the
autocrine agent) that binds to autocrine receptors on
the same type of cell, leading to changes in the cells.

This can be contrasted with paracrine signaling,


intracrine signalling, or classical endocrine signaling.
OVERVIEW OF SIGNAL
TRANSDUCTION PATHWAYS
Signal Transduction
• In biology, signal transduction refers to any
process by which a cell converts one kind of
signal or stimulus into another. Most processes
of signal transduction involve ordered
sequences of biochemical reactions inside the
cell, which are carried out by enzymes and
activated by second messengers, resulting in a
signal transduction pathway.
Such processes are usually rapid, lasting on the order of
milliseconds in the case of ion flux, or minutes for the
activation of protein and lipid-mediated kinase cascades,
but some can take hours, and even days (as is the case
with gene expression), to complete.
The number of proteins and other molecules participating
in the events involving signal transduction increases as
the process emanates from the initial stimulus, resulting
in a "signal cascade," beginning with a relatively small
stimulus that elicits a large response.
This is referred to as amplification of the signal.
Signaling molecules
Most signal transduction involves the binding of extracellular signaling
molecules (or ligands) to cell-surface receptors that face outward
from the plasma membrane and trigger events inside the cell. Also,
intracellular signaling cascades can be triggered through cell-
substratum interactions, as in the case of integrins, which bind
ligands found within the extracellular matrix. Steroids represent
another example of extracellular signaling molecules that may cross
the plasma membrane due to their lipophilic or hydrophobic
nature.
Many, but not all, steroids have receptors within the cytoplasm, and
usually act by stimulating the binding of their receptors to the
promoter region of steroid-responsive genes.
Within multicellular organisms, there is a diverse number of small
molecules and polypeptides that serve to coordinate a cell's
individual biological activity within the context of the organism as a
whole.
These ligand molecules have been functionally
classified as:
• Hormones (e.g., melatonin),
• Growth factors (e.g. epidermal growth factor),
• Extra-cellular matrix components (e.g., fibronectin),
• Cytokines (e.g., interferon-gamma),
• Chemokines (e.g., CCL5 or RANTES = Regulated upon
Activation, Normal T-cell Expressed, and Secreted),
• Neurotransmitters (e.g., acetylcholine), and
• Neurotrophins (e.g., nerve growth factor).
CCL5 is an 8kDa protein classified as a chemotactic cytokine or
chemokine. CCL5 is chemotactic for T cells, eosinophils, and
basophils, and plays an active role in recruiting leukocytes into
inflammatory sites.

With the help of particular cytokines (i.e., IL-2 and IFN-γ) that are
released by T cells, CCL5 also induces the proliferation and
activation of certain natural-killer (NK) cells to form CHAK (CC-
Chemokine-activated killer) cells. It is also an HIV-suppressive
factor released from CD8+ T cells. This chemokine has been
localized to chromosome 17 in humans.

RANTES was first identified in a search for genes expressed "late" (3–
5 days) after T cell activation. It was subsequently determined to be
a and expressed in more than 100 human diseases. RANTES
expression is regulated in T lymphocytes by Kruppel like factor 13
(KLF13).
1PMI human rantes
sekian
Lihat: Medicinal Chemistry and Drug Discovery

S-ar putea să vă placă și