You are on page 1of 24

TARGETING RECEPTOR

TYROSINE KINASE
WHAT IS TYROSINE KINASE?

• A tyrosine kinase is an enzyme that can transfer a phosphate group from ATP to a tyrosine
residue in a protein.
• Tyrosine kinases are divided into 2 main groups:
1. The receptors: they posses an intrinsic tyrosine kinase activity that is part of the receptor
protein
2. The non-receptors: are receptors lacking self-contained kinase function recruit activities on
intracellular protein kinases to the plasma membrane
WHAT ARE RECEPTOR TYROSINE KINASES?

• Receptor tyrosine kinases (RTKs) are cell surface receptors that play an essential role in signal
transduction from extracellular stimuli. Receptor tyrosine kinases are transmembrane proteins,
which consist of several domains that are activated upon ligand binding to their extracellular
regions, triggering downstream signaling cascades. They are involved in various regulatory
processes, such as cell survival, growth, differentiation, adhesion, proliferation, and motility.
Impaired gene functions by mutations or deletions may cause the abnormal expression of
protein kinases, which, in turn, entails tumor formation and progression
DOMAINS OF RTK

1. Extracellular ligand binding domain


2. Intracellular tyrosine kinase domain, with amino acid sequences in ATP binding and
substrate binding regions
3. Intracellular regulatory domain
4. Transmembrane domain
HOW ARE RTKS ACTIVATED?

an un-activated RTK receptor encounters a ligand. Upon binding, the receptor forms a complex of
proteins that phosphorylate each other. In turn, this phosphorylation affects other proteins in the cell
that change gene transcription.
RTK’S & DRUG DESIGN

• Rational drug design is accomplished through the complementary use of structural biology and
computational biology of biological macromolecules involved in disease pathology. Most of
the known theoretical approaches for drug design are based on knowledge of the biological
targets to which the drug binds. This approach can be used to design drug molecules that
restore the balance of the signaling pathway by inhibiting or stimulating biological targets by
molecular modeling procedures as well as by molecular dynamics simulations. Type III
receptor tyrosine kinase affects most of the fundamental cellular processes including cell cycle,
cell migration, cell metabolism, and survival, as well as cell proliferation and differentiation.
Many inhibitors of successful rational drug design show that some computational techniques
can be combined to achieve synergistic effects.
RECEPTOR TYROSINE KINASES

• RTKs as well as nonreceptor tyrosine kinase play a critical role in development and cell
growth, and a wealth of information has accumulated implicating deregulated and
dysfunctional RTKs in cancer and other diseases. Aberrant RTK activation in human cancers is
mediated by four principal mechanisms: autocrine activation, chromosomal translocations,
RTK overexpression, and gain-of-function mutations.
• Sequencing efforts in a wide variety of tumors have identified numerous mutations in RTKs.
Consequently, the focus has been to develop RTK antagonists, rather than agonists which
promote cell proliferation and survival. Nonetheless, the discovery and development of RTK
antagonists have lagged behind the discovery and development of agents that target G-protein-
coupled receptors.
RTK’S & CANCER

• Recent advances have implicated the role of tyrosine kinases in the pathophysiology of cancer.
Though their activity is tightly regulated in normal cells, they may acquire transforming
functions due to mutations, overexpression and autocrine paracrine stimulation, leading to
malignancy. Constitutive oncogenic activation in cancer cells can be blocked by selective
tyrosine kinase inhibitors and thus considered as a promising approach for innovative genome
based therapeutics.
ONCOGENIC ACTIVATION OF TYROSINE KINASE

• Normally the level of cellular tyrosine kinase phosphorylation is tightly controlled by the
antagonizing effect of tyrosine kinase and tyrosine phosphatases. There are several mechanisms
by which tyrosine kinase might acquire transforming functions, but the ultimate result is the
constitutive activation of normally controlled pathways leading to the activation of other
signaling proteins and secondary messengers which serves to hamper the regulatory functions
in cellular responses like cell division, growth and cell death. Constitutive activation of
tyrosine kinase may occur by several mechanisms.
TYROSINE KINASE INHIBITORS

• Kinase inhibitors are now one of the major categories of chemotherapy medicine.  Over 45
kinase inhibitors are approved in the US for cancer treatment with more under development.
Kinases are a class of enzyme that promote phosphorylation. When these kinases go wrong,
normal cellular function can go awry. Kinase deregulation can contribute to the growth of
cancer. Drugs are given to stop kinases can slow the proliferation of malignant cells and
angiogenesis (growth of blood vessels). 
TYROSINE KINASE INHIBITORS

• Receptor Tyrosine Kinases are a family of tyrosine protein kinases. RTKs span the cell
membrane with an intracellular and extracellular portion. The intracellular portion removes a
phosphate group, a process called dephosphorylation, from the coenzyme messenger ATP. The
extracellular portion has sites to which signal sending proteins and hormones can bind. Many
of these signaling binders are growth factors.
GROWTH FACTORS

• Growth factors are involved in the initialization and regulation of cell cycles. The type of
growth factor determines its effects on the cell. There are three primary growth factors that
relate to tyrosine kinase. The receptors of these growth factors are members of the RTK
family. 
1. Epidermal growth factors (EGF) help regulate cell growth and differentiation. 
2. Platelet-derived growth factors (PDGF) regulates cell growth and development.
3.  Vascular endothelial growth factors (VEGFR) are involved in the creation of blood
vessels.
GROWTH FACTORS

• The growth factors, and the kinases, act together as though they are attached to an “on/off” switch. The
removal of a phosphate group changes the shape and actions of the protein. This essentially “turns on”
the cellular action (or actions). When the cellular action(s) is completed, the phosphate group is removed
and that protein is “turned off.” This “on/off” process can become disrupted, often by a mutated kinase,
and actions can become unregulated. An unregulated RTK bound to EGF, for example, could lead to
uncontrolled growth and division in the cell. The rapid cell growth could then lead to cancer. Mutations
of RTKs often lead to oncogenes, which are genes that help turn a healthy cell into a cancerous cell.
• Tyrosine kinase inhibitors treat cancer by correcting this deregulation. Imatinib, for example, blocks a
kinase receptor from binding to ATP, preventing the phosphorylation that would benefit the cancerous
cell and promote cell division. Gefinitib inhibits EGFRs, preventing that signal from being stuck “on”
and creating uncontrolled proliferation.
TYROSINE KINASES AS TARGETS FOR ANTICANCER
AGENTS

• The role of tyrosine kinases in cancer molecular pathogenesis is immense and recently kinases
have come in vogue as potential anticancer drug targets, as a result a couple of anticancer drugs
are in the market. The complexity and the number of tyrosine kinases have greatly increased
with the sequencing effort of the Human Genome Project, thus providing more opportunities
for drug discovery. Recent understanding of the molecular pathophysiology of cancer have
highlighted that many tyrosine kinases are found upstream or downstream of epidemiologically
relevant oncogenes or tumor suppressor, in particular the receptor tyrosine kinases
RTKS AS DRUG TARGETS

• Several drugs have been developed for the treatment of cancers and other diseases caused by
aberrant RTK activation.
• These drugs fall into two categories:
1. small-molecule tyrosine kinase inhibitors (TKIs) that target the ATP-binding site of the
intracellular TK domain
2. monoclonal antibodies that both interfere with RTK activation and target RTK-expressing
cells for destruction by the immune system
RTKS AS DRUG TARGETS

• Two generations of small-molecule TKIs targeting RTK as well as nonreceptor TK have been
approved for the treatment of human malignancies since the breakthrough with imatinib (is an
oral chemotherapy medication used to treat cancer)

1. First-generation TKIs like imatinib bind to their target, the catalytic site in the TK domain,
through classic competitive binding with ATP, and are fairly selective for their respective TK
targets.
FIRST-GENERATION TKIS- IMATINIB

• mechanism of action: binds to a segment of the kinase domain that fixes the enzyme in a closed or
nonfunctional state, in which the protein is unable to bind its substrate/phosphate donor, which is ATP.
• Pharmacokinetics:
1. Absorption: oral bioavailability- 98%
2. Distribution: highly protein bound
3. Metabolism: primarily by CYP3A4 (is an important enzyme in the body, mainly found in the liver and
in the intestine. It oxidizes xenobiotics (small foreign organic molecules), such as toxins or drugs, so
that they can be removed from the body.)
4. Elimination: fecal- 65-80% / renal – 10-13%
 half life: 18 hours
FIRST-GENERATION TKIS - IMATINIB

 Imatinib blocks the ATP binding center of Bcr-Abl (which protein that has role in the oncogenic
pathway) thus inhibiting its phosphorylation activity. Therefore CML won’t be produced (Chronic
myelogenous leukemia (CML) is an uncommon type of cancer of the bone marrow. CML causes an
increased number of white blood cells in the blood.)
SECOND GENERATION TKIS

2. Second generation of TKIs is now emerging and being introduced into clinical trials. The
two most commonly employed strategies are introducing covalent (irreversible) binding of
the drug to the RTK target, and kinase multitargeting by broadening the affected RTK targets
of the drug within the cell. These novel TKIs appear promising as cancer treatments, and
these drugs attempt to improve upon their first-generation predecessors.
SECOND GENERATION TKIS-
NILOTINIB
• Imatinib has benefited a large number of patients with CML. However, imatinib resistance has
emerged as an important clinical challenge. Novel treatment strategies have been investigated
after failure of imatinib therapy. The availability of highly potent tyrosine kinase inhibitors,
such as nilotinib, has broadened the treatment armamentarium in CML. Nilotinib appears to
overcome imatinib resistance in patients with chronic, accelerated, and blastic phase CML,
producing sustained cytogenetic and hematological responses. Combination strategies may be
useful, although these have not yet been examined in clinical studies. With the availability of
nilotinib, treatment options are increasing in CML, and this is likely to continue in the future.
SECOND GENERATION TKIS- NILOTINIB
• Indication: it is used in the treatment of chronic phase and accelerated phase of Chronic
myelogenous leukemia in adult patients who are resistant to imatinib
• Pharmacokinetics:
1. Absorption:
 peak plasma levels-3 hours
 Approximately 30% of an oral dose of nilotinib is absorbed after administration
 Fatty meals food increases absorption
2. Distribution:
 highly protein bound
 Plasma concentration reaches a steady state only after 8 days of daily dosing
CONCLUSION

• In conclusion, the role of tyrosine kinase in the control of cellular growth and differentiation is
central to all organisms and has been found to participate in human neoplastic diseases. Tyrosine
kinase inhibitors and their potential in clinical application are well documented by dramatic
examples like, Gleevec, Iressa and Herceptin. Several tyrosine kinase inhibitors are undergoing
human trials and several are in the pipeline of drug discovery. The activities of these drugs are
restricted to cancers with alterations in kinase targets, hence broad application of this treatment
strategy are challenging. The quick selection of epidemiologically relevant, drug-able tyrosine
kinase targets coupled to efficient lead finding and optimization needs more intervention in the
area of high throughput cancer genome based molecular therapeutics. All these concerted effort
may pave the silver lining to tailor made personalized cancer therapeutics.
REFERENCES

1. https://www.nature.com/scitable/topicpage/rtk-14050230/
2. https://molecular-cancer.biomedcentral.com/articles/10.1186/s12943-018-0782-4
3. Textbook of drug design and discovery 5th edition
4. https://chemoth.com/types/kinaseinhibitors
5. https://www.intechopen.com/books/leukemia/modern-therapy-of-chronic-myeloid-leukemia
6. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1074718/
7. https://pubmed.ncbi.nlm.nih.gov/29932031/

You might also like