Hallmarks of Cancer

Download as pptx, pdf, or txt
Download as pptx, pdf, or txt
You are on page 1of 21

HALLMARKS

OF CANCER
Submitted To- Submitted By-
Dr. Ausaf Ahmad Ananya Singh
Assistant Professor A7104120008
AIB, AUUP B.Tech Bt (V)
Lucknow Campus
INTRODUCTION
Cancer is the uncontrolled growth of abnormal cells anywhere in the body. These abnormal
cells are termed cancer cells, malignant cells, or tumor cells. These cells can infiltrate
normal body tissues.
In other words, Cancer is a disease that results when cellular changes cause the
uncontrolled growth and division of cells.
Over the past several decades, hundreds of genes that are mutated in cancer have been
discovered and were thoroughly researched upon to describe the functional alterations that
result in cancer.
However, the avalanche of genes identified over the last decades has limited the scope of
understanding the underlying characteristics of cancer gene by gene.
Examining the common phenotypic traits that are shared by cancer cells as result of their
genetic and epigenetic modification is a far more manageable way to understand the
biology of cancer.
HALLMARKS OF CANCER
Over the past few decades researchers have discovered several molecular, biochemical, and cellular
characteristics—acquired abilities—that are shared by the majority, and possibly all, kinds of human
cancer. 
It appears that all cancers display eight fundamental changes in cell physiology, which are considered
the ‘hallmarks of cancer’. These changes are:
1. Self-Sufficiency in Growth Signals: Oncogenes
2. Insensitivity to Growth Inhibition: Tumour Suppressor Genes
3. Altered cellular metabolism
4. Evasion of apoptosis
5. Limitless replicative potential
6. Sustained angiogenesis
7. Ability to invade and metastasize Hallmarks of cancer (Adapted from Hanahan
D, Weinberg RA)
8. Ability to evade the host immune response
01 Self Sufficiency in Growth Signals: Oncogenes
Proto-oncogenes are a group of typical genes in cell that have a key role in cell division and
cell prolifiration. The proto-oncogenes encode for growth factor, growth factor receptor,
signal transducers, transcription factors and cell cycle regulators.
Oncogenes are the mutated genes that promote autonomous cell growth in cancer cells.
These mutated genes, i.e. oncogenes encode for oncoproteins that resemble the normal
product of proto-oncogenes with the most important difference being their constitutive
activity. As a result of this constitutive activity, oncoproteins endow cells with self
sufficiency in growth.

Gain of Function
Proto-oncogenes Oncogenes
Growth Factors Growth Factor Receptor
Normal cells require stimulation by Overexpression of genes encoding for
growth factors to proliferate. Most growth factor receptor in cancer cells
growth factors are made by one type of can render cancer cells hyper-responsive
cells and act on other types of to levels of growth factor that normally
neighboring cells i.e paracrine action. would not trigger proliferation.
However, some cancer cells acquire the For example, EGFR is overexpressed in
ability to synthesize the same growth 80% of squamous cell carcinomas of the
factor to which they are responsive lung, 50% of glioblastomas and almost
creating an autocrine loop. The signals all epithelial tumors of head and neck.
transduced by oncoproteins cause
overexpression and increased secretion
of growth factors, thereby amplifying
the autocrine loop and causing
excessive proliferation.
Signal Transducers

RAS family genes (HRAS, KRAS, NRAS) that encode for RAS family proteins, that are GTP binding,
membrane associated G-Proteins that act as signal transducers.
Constitute the most common type of abnormalities involved in proto-oncogenes in human tumors. Apx. 15-
30% of all human tumors express mutated RAS genes.
Let us first understand the activity of RAS protein in normal cells

Receptor Exchange of Activate Stimulate MAPK and


Growth factors
Tyrosine Kinase GTP RAS P13/AKT pathway

RAF/MEK/ERK pathway and PI3K/AKT pathways are involved in cell growth, proliferation and survival.

Now, any mutation in the RAS gene, that renders it constitutively active (GTP-bound state), or any activating
mutation in the growth factor receptor that act upstream of RAS or any aberrant RAS effector activation that
increases the signalling mechanism of RAS may lead to development of cancer.
02 Insensitivity to Growth Inhibition: Tumour
Suppressor Genes
Tumor Suppressor Genes apply breaks to cell proliferation and thus checks cancer in normal cells.
They are recruited at G1/S and G2/M check points of cell cycle and are responsible for halting the
cell cycle in case of any abnormality.
Any abnormalities in these genes, lead to failure of growth inhibition and incipient cancer cells
evade the antiproliferative signals.
The loss of function of tumor suppressor genes contributes to unregulated cell growth and
proliferation.
TP53 (aka ‘guardian of genome’ and ‘master watchman’) is a major tumor suppressor gene that
encodes for p53 protein that regulates cell cycle progression, DNA repair, cellular senescence and
apoptosis)
The frequent loss of p53 functions in human tumor reflects its critical role in cancer development.
Loss of function mutation in TP53 are found in more than 50% of cancers.
No DNA Damage No DNA Damage
MDM2
MDM2 P
p53 p53

MDM2 p21 p21


Cyclin-CDK
p53
Complex
Inhibition of cyclin-CDK complex
Binding of MDM2 with p53 causes cell cycle arrest
causes ubiquitin mediated If the cell rectifies the DNA damage, it is allowed to undergo further cell
proteasomal degradation of division. However, in case it is expensive to repair the DNA, p53
p53 activates various apoptotic transcription factor and other apoptotic
effectors

With loss of function of p53, cell proliferation is not efficiently regulated and DNA
damage can accumulate in cells.
These cells can then continue to divide out of control, leading to tumor growth.
Fortunately, we have 2 copies of TP53 gee, one from each parent.
As mutations are recessive, both the alleles of TP53 needs to be corrupted to lose
normal TP53 function.
Less commonly, individuals inherit one mutated TP53 allele. Such individuals are said
to have Li-Fraumeni syndrome and are at a 25% greater chance of developing a
malignant tumour by age 50 than general population.
The spectrum of tumour that develop in individuals with Li-Fraumeni syndrome is
quite varied, with most common types being, sarcomas, breast cancer, leukaemia and
brain tumours.
03 Altered cellular metabolism
Even in the presence of ample oxygen, cancer cells demonstrate a dinstinctive form of cellular
metabolism characterized by high levels of glucose uptake and increased conversion of glucose
to lactose via glycolytic pathways. This phenomenan is called ‘warburg effect’ aka aerobic
glycolysis.
However, this phenomeana is not cancer specific, it is aso observed in embryonic tissues, but it
becmes “fixed” cancer cells.
Aerobic glycolysis provides rapidly dividing tumor cells with metabolic intermediates that are
needed for synthesis of cellular components.
In Warburg effect metabolic reprogramming is produced by signaling cascades downstream of
growth factor receptors, the very same pathways that are deregulated by mutations in oncogenes
and tumor suppressor genes in cancers.
Crosstalk between pro-growth signaling factor and cellular metabolism

P13/AKT Signaling Receptor Tyrosine MYC


Kinase
Upregulates activity of Rapidly dividing cells, both normal and Pro-growth pathways
glucose transporters and malignant express M2 isoform of upregulate expression of TF
multiple glycolytic pyruvate kinase, which catalyzes the MYC, which drives changes
enzymes, which in turn conversion of phosphoenolpyruvate to in gene expression that
increases glycolysis; pyruvate. support anabolic
promotes shunting of metabolism and cell growth.
mitochondrial intermediates Receptor tyrosine kinase phosphorylate
to pathways leading to lipid the m2 form of pyruvate kinase that
biosynthesis; and stimulates attenuates its enzymatic activity that
factors that are required for leads to build up of upstream glycolytic
protein synthesis. intermediates that gets used for synthesis
of DNA, RNA and protein required for
cell growth.
04 Evasion of apoptosis
Apoptosis i.e programmed cell death is a protective
response to several pathologic conditions that might
contribute to malignancy if cells remained viable.
Through apoptosis, any cell with genomic injury can be
induced to die, eliminating the chance that such cell might
go on to give rise to neoplasm.
Apoptosis can be initiated through both extrinsic (signaling
through death receptor) and intrinsic pathways (DNA
damage stress, injure, hypoxia), both of which result in
activation of a proteolytic cascade of caspases that destroy
the cell.
Abnormalities of both pathways are found in cancer cells,
but abnormalities of intrinsic pathways are more commonly
found in cancer cells. Intrinsic and Extrinsic Pathways of
Apoptosis
Apoptosis in normal cell is guided by cell death receptor, CD95, other genes regulating
apoptosis and cancer pro-apoptotic factors like BAD, BAX, BID and p53 and apoptosis
inhibitors like BCL-2, BCL-X.
In cancer cells, the function of apoptosis is interfered due to mutations in the above genes
which regulate apoptosis in normal cells.
Example:
 BCL-2 gene mutation removes apoptosis-inhibitory control on cancer cells, thus more live
cells undergo mitosis contributing to tumor growth. In apx. 85% B-cell lymphomas, the
anti-apoptotic gene BCL-2 is overexpressed due to translocation.
 CD95 receptors are depleted in hepatocellular carcinoma and hance tumor cells escape
apoptosis.
 MYC oncogene and p53 tumour suppressor gene are also involved in apoptosis. While
MYC allows cell growth, BCL-2 inhibits cell death; thus MYC and BCL-2 together allow
cell proliferation. Normally, p53 activates pro-apoptotic gene BAX but mutated p53
reduces apoptotic activity and thus allows cell proliferation.
05 Limitless Replicative Potential
All cancer contains cells that are immortal and have limitless replicative potential.
Some cell lines established from cancers have now been proliferating ceaselessly in laboratories for
more than 60 years.
Though the actual mechanism through which cancer cells achieve immortality is not fully understood.
But three interrelated factors appear critical to the immortality of cancer cells. They are:
1. Evasion of senescence:
Most normal human cells have the capacity to divide 60-70 times. After this cells leave cell cycle
permanently i.e become senescent.
Senescent is associated with upregulation of tumor suppressor genes.
These genes are believed to contribute to senescence by maintaining Rb in hypo-phosphorylated
state, which favors cell cycle arrest.
This Rb-dependent G1/S cell cycle checkpoint is apparently disrupted in all cancers by a wide
variety of acquired genetic and epigenetic aberrations.
2. Evasion of mitotic crisis: 3. The capacity for self renewal:
While cells that are resistant to senescence have At least some cells in all cancers must be stem cell-
increased replicative capacity, they still are not like. These cells are sometimes referred to as
immortal as they eventually enter into a phase cancer stem cells. These may arise through
referred to as “mitotic crisis’ that is ascribed to transformation of normal stem cell or through
progressive shortening of telomere. acquired genetic lesions that impart a stem-like
Telomerase, an enzyme that is required for state to more mature cells.
maintenance of telomere is expressed at very low In chronic myelogenous leukemia (CML), the
levels in most somatic cells and thus, after repeated BCR-ABL gene ingrains self renewal capacity to
cell cycles, even if cell evades senescence, the hematopoietic stem cells.
telomere is eroded which is sensed as DNA damage Acute myeloid leukemia (AML) have shown that
by apoptotic factors and the cell enters apoptosis. cancer stem cells in this disease arise from more
However, if in case of crisis, cells reactivate differentiated hematopoietic progenitors that
telomerase, they can restore their telomere and acquire an abnormal capacity for self-renewal.
survive. Such cells are at a high risk for malignant PML-RARA protein is associated with AML
transformation.
Telomere maintenance is seen in virtually all types
of cancers, and in 85-95% cases this is due to
upregulated telomerase expression.
06 Sustained Angiogenesis
Like normal tissues, tumors require sustenance in the form of nutrients and oxygen as well as an ability to
evacuate metabolic wastes and carbon dioxide.
The tumor associated neovascularization, generated by the process of angiogenesis addresses these needs.
Normally, angiogenesis is controlled by a balance between angiogenesis promoters and inhibitors; in
tumors this balance is skewed in favor of promoters.
The local balance of angiogenic and anti-angiogenic factors is influenced by several factors:
1. Relative lack of oxygen stabilizes HIF, which then activates the transcription factors that activates
pro-angiogenic cytokines VEGF and b-FGF. This factors stimulate the proliferation of endothelial
cells and guides growth of new vessels towards tumors.
2. P53 also stimulate expression of anti-angiogenic factors such as thrombospondin-1 and repress
expression of pro-angiogenic factors such as VEGF. Thus loss of p53 function also provides a more
permissive environment for angiogenesis.
3. Gain of function mutations in RAS and MYC also upregulate the production of VEGF.
Invasion and metastasis are the results of
complex interactions between cancer
cells and normal stroma and are the
07 major causes of cancer-related morbidity
and mortality.
Ability to The invasive-metastatic cascade involves
two phases:
metastasize and 1. Invasion of extracellular matrix
invasion (ECM)
2. Vascular dissemination, homing and
colonization of tumor cells
1. Invasion of extracellular matrix (ECM)
Tumor cells must first penetrate the ECM that includes basement membrane and
interstitial connective tissue.

i. Loosening of intercellular ii. Degradation of ECM iii. Attachment of tumor cells iv. Migration and invasion of
Junctions Degradation of basement to ECM proteins tumor cells
Dissociation of cancer cells membrane and interstitial Cleavage of basement This is stimulated and directed
from one other is often result of connective tissue is second step membrane proteins, collagen by tumor derived cytokines
alteration in intercellular in invasion. Tumors do this IV and laminin by MMPs such as autocrine motility
adhesion molecule. either by secreting proteolytic generate novel sites that bind factor.
Cadherin mediate the adhesion enzymes or by inducing stromal to receptors on tumor cells and
of cells. Presumably, the cells to elaborate proteases stimulate migration.
function of cadherin is lost in (like cathepsin D, matrix
cancer cells. metalloproteases (MMP)
2. Vascular Dissemination and Homing of Tumor Cells
Once in circulation, tumor cells tend to aggregate in clumps. This is favored homotypic adhesion
among tumor cells as well as heterotypic adhesion between tumor cells and blood cells, typically
platelets. This platelet-tumor aggregates enhance tumor cell survival and implantability.
The anatomic location and vascular drainage of the original tumor influence where circulating
tumor cells leaves the capillaries to form secondary deposits.
Most metastases develop in the first capillary bed that is available to the tumor. However, certain
tumor have a preference for certain organs. This is known as “organ tropism”.
This organ tropism may be related to:
• Tumor cells may have adhesion molecules whose ligands are preferentially on endothelial cells
of target organ.
• Chemokines have an important role in determining the target tissues for metastasis.
• Some tissues may have a non-permissive environment. Example, spleen and skeletal muscle
are highly vascular, but are rarely the site of metastasis.
08 Evasion of Host Immune Response
Tumor cells can be recognized by the immune system as non-self
and destroyed.

It is clear that tumor cells must develop mechanisms to escape the


immune system in immunocompetent hosts. These mechanisms
include:

i. Selective outgrowth of antigen-negative variants

ii. Loss or reduced expression of MHC molecules

iii. Activation of immunoregulatory pathways

iv. Secretion of immunosuppressive factors by cancer cells.

v. Induction of regulatory T cells.


Thank You!

You might also like