0% found this document useful (0 votes)
22 views87 pages

Understanding Cancer Genetics and Causes

Uploaded by

yisakbedlu
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PPTX, PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
22 views87 pages

Understanding Cancer Genetics and Causes

Uploaded by

yisakbedlu
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PPTX, PDF, TXT or read online on Scribd

Cancer Genetics

By: Amanuel W. (2023)


Outlines:-
• Introduction to cancer

• Relationship between cell division, apoptosis and cancer

• Oncogens and Oncogenes

• Tumor suppresssor genes

• Tumor markers
Introduction to cancer

 The term “cancer” is derived from Latin word “cancrum” or Greek “karkinoma”,

which means “crab”. The disease is so called because of swollen veins around the

area, resemble a crab's limbs.


Introduction to cancer
Indian Medical Science had identified cancer, gave the name “arbuda”, which

literally means the number 108(ten the power of 8) , identifying the extreme

cellularity of the cancer tissue.

 The International Union Against Cancer (UICC; Union Internationale Contre le

Cancer) has defined cancer as a disturbance of growth characterized by excessive

proliferation of cells without apparent relation to the physiological demands of the

organs involved.

Oncology:- deals with the etiology, diagnosis, treatment, prevention and research

aspects of cancer
Intro….

• It begins due to abnormal changes in a single cell’s genetic material that affect the

mechanisms of normal cell growth, regulation, and cell death, which finally leads to

uninhibited cell growth.

• When broad classes of genes(proto-oncogenes, tumor suppressor genes, DNA synthesis

and repair genes, genes regulating apoptosis, or genes causing evasion of immune

surveillance) mutated, it cause gain- or loss-of-function or misregulation, and can thus

result in the development of a tumor.

• Cancer is of clonal origin, with a single abnormal cell (often with many genetic alterations)

multiplying to become a mass of cells forming a tumor.


Intro ….
• Genetic damage causing cancer can be due to acquired, or inherited mutations. Aquired

mutations occur either by errors in DNA replication or DNA repair, and are termed replication

mutations (R), or by exposure to environmental carcinogens, and are termed environmental

mutations (E) (radiation, chemicals and viruses).

• The third major class of oncogenic mutation are termed hereditary mutations (H); inherited

mutations are derived from one or both parents. Such hereditary abnormalities result in a

number of familial conditions that predispose to hereditary cancer. These mutations are found

in specific genes (eg, tumor suppressor genes; DNA repair genes, cell cycle control genes, etc)

present in the germ cells.

• Mutations of the R, E, and H variety collectively cause the majority of human cancers, though the

exact percentage of a particular type of cancer caused by these three types of mutations varies.
Intro…
 In most cases abnormal changes are caused by combination of both hereditary
factors (inheritance of one or more genetically inactive proteins) and
environmental factors such as physical carcinogens (e.g. ionizing radiation),
biological carcinogens (e.g., oncogenic viruses) and chemical carcinogens (e.g.
components of tobacco smoke, asbestos, and radon).

 Cancer is a rapidly growing disease and the leading cause of death globally, which
was responsible for almost 9.7 million deaths and 20 million new cases in 2022. One
of six deaths in the world is because of cancer.
RADIATION, CHEMICALS, AND CERTAIN VIRUSES ARE MAJOR KNOWN CAUSES OF
CANCER
In general, there are three classes of environmental carcinogens that can induce
tumor formation: radiation, chemicals, and certain oncogenic viruses.
The first two carcinogens cause mutations in DNA, while viruses generally act by
introducing novel genes into normal cells.
Radiation can be Carcinogenic
• Ultraviolet rays (UV), x-rays, and γ-rays are mutagenic and carcinogenic. These
agents damage DNA in a number of ways.
• Mutations in DNA, if not corrected, are thought to underlie the carcinogenic effect of
radiation, although the exact pathways are still under investigation.
• Additionally, x-rays and γ-rays can induce formation of reactive oxygen species
(ROS), which as noted above are also mutagenic and probably contribute to the
carcinogenic effects of radiation.
Many Chemicals are Carcinogenic
Most chemical carcinogens are thought to covalently modify DNA thereby forming a
wide range of nucleotide adducts.

Depending on the extent of damage to DNA and its repair by DNA repair systems, a
variety of mutations in DNA can result from exposure of an animal or human to
chemical carcinogens, some of which contribute to the development of cancer.

Some chemicals interact directly with DNA (eg, mechlorethamine and β-


propiolactone), but others, termed procarcinogens, require conversion by enzyme
action to become ultimate carcinogens.

Conversion of chemicals to ultimate carcinogens is principally due to the actions of


various species of cytochrome P450 located in the endoplasmic reticulum (ER).
Mechanisms of action of chemical carcinogens are:

(a) Carcinogens are generally electrophiles (molecules deficient in electrons); they readily attack

nucleophilic (electron rich) groups of DNA.

(b) Carcinogens may bind covalently to cellular DNA. N2, N3, and N7 atoms of guanine are highly

prone to addition of carcinogen groups.

(c) These changes will lead to DNA alterations, in spite of DNA repair, with increased probability of

mutations.

Chemical carcinogens may produce the cancer:

(a) At the site of exposure, e.g. buccal cancer in tobacco chewers, skin cancer in tar workers.

(b) At the site of metabolism, e.g. liver cancer produced by aflatoxin.


Many Chemicals are Carcinogenic

Chemical carcinogenesis involves two stages—initiation and promotion.

Initiation: is the stage where exposure to a chemical cause genomic DNA

damage, some of which go unrepaired, and is a necessary initial event for a

cell to become cancerous.

Promotion: is the stage at which an initiated cell begins to grow and

proliferate abnormally.

The cumulative effect of these two events is a neoplasm.


Many Chemicals are Carcinogenic
Chemical carcinogens can be identified by testing for their ability to induce mutations. A simple way to
do this is by using the Ames assay.

This relatively simple test, which detects mutations in the bacterium Salmonella typhimurium caused
by chemicals, has proven very valuable for screening purposes.

A refinement of the Ames test is to add an aliquot of mammalian ER to the assay, to make it possible to
identify procarcinogens.

Very few, if any, compounds that have tested negative in the Ames test have been shown to cause
tumors in animals. However, animal testing is required to show unambiguously that a chemical is
carcinogenic.

The chemical tested will increase the frequency of reversion of His- to His+ cells if it is a mutagen and,
therefore, a potential carcinogen. A control plate (not shown) contains the solvent in which the
suspected mutagen is dissolved.
Fig: The Ames assay to screen for mutagens
Many Chemicals are Carcinogenic
It should be noted that compounds that alter epigenetic factors (such as DNA methylation
and/or histone posttranslational modifications); that might predispose to cancer, would
not test positive in the Ames test, as they are not mutagenic.

These may be introduced into the body by means of

(a) Occupation (aniline, asbestos)

(b) diet (aflatoxins) or

(c) Lifestyle (smoking).

Chemical carcinogens act cumulatively. Tobacco, food additives, coloring agents, and
aflatoxins are common carcinogens in our environment
 Methyl cholanthrene is a powerful carcinogen,
only nanograms are sufficient to produce a
tumor in a mouse

 Aflatoxins: are a group of chemically related


compounds synthesized by the fungi,
Aspergillus flavus. The mould grows on rice,
wheat and groundnut, when kept in damp
conditions. The fungi may grow in cattle fodder,
which may enter into human body through the
cow's milk. Aflatoxins are powerful carcinogens,
which produce hepatomas.
Fig: Powerful chemical carcinogens
Cigarette
Lung cancer is associated with the habit of cigarette smoking. Cigarette contains many
carcinogens, the most important group being benzo(a)pyrenes.

Other important deleterious substances in cigarette smoke are nicotine, carbon monoxide,
nitrogen dioxide and carbon soot.

Statistically, it is estimated that one cigarette reduces 10 minutes from the life span of the
individual.

The incidence of lung cancer is increased to 15 times more in persons smoking 10 cigarettes per
day and 40 times more when smoking 20 cigarettes per day.

 Alcohol intake increases the risk of oral, pharyngeal, esophageal and liver cancers.

Diet high in total fat and cholesterol, increases the risk of colon, breast and prostate
cancers.
Promoters of Cancer
 Most carcinogens require promoters for the production of a cancer.

 Benzopyrene applied on skin does not produce cancer. Croton oil application also
does not lead to skin cancer.

 But when benzopyrene application is followed by croton oil, tumor is developed. In this
case, croton oil is termed as the promoter.

 The active agent in croton oil is a phorbol ester, tetradecanoyl phorbol acetate (TPA).
It activates Protein kinase-C.

 This results in phosphorylation of membrane proteins, leading to the triggering of


malignancy. The carcinogen produces a mutation, but the promoter gives the drive
for unchecked cell division.
Progression
The biological history of a tumor shows progression of malignancy. Cells with faster growth
rate have a selection advantage. Thus cells with increased malignant character are
progressively selected.

Development of familial adenomatous polyposis is a good example for multistep


progression.

Mutations in the APC gene are inherited from parents. By the time the patient becomes adult,
there will be different dysplastic aberrant crypts in the large intestine.

Some of the cells will get somatic mutations in the K-Ras gene; these will progress to form
adenomas.

Further mutation in TGF gene or p53 gene or Bax gene will give the push for the development
of malignancy.
Antimutagens
i. These are substances which will interfere with tumor promotion. Vitamin A and
carotenoids are shown to reverse precancerous conditions.
ii. Vitamin E acts as an antioxidant, preventing the damage made by free radicals and
superoxides.
iii. Vitamin C regularly given to persons working with aniline prevented the production
of new cancer cases.
iv. Tubers, beans and leafy vegetables are shown to interrupt tumor promotion.
v. Curcumin, the yellow substance in Turmeric is known to prevent mutations.
vi. The dietary fiber has also beneficial effect. Low protein, low fat, diet decreases the
risk of cancer in animal studies.
Certain Human Cancers are caused by Viruses
• Both DNA and RNA viruses have been identified as being able to cause cancers in humans.

• In general, the genetic material of viruses is incorporated into the genome of the host cell.

• In the case of RNA viruses, this would occur after reverse transcription of the viral RNA to

viral DNA. Such integration of viral DNA (called the provirus) into the host DNA results in

various effects such as deregulation of the cell cycle, inhibition of apoptosis, and

abnormalities of cell signaling pathways.

• DNA viruses often act by downregulation of the expression, and/or function of tumor

suppressor genes P53 and RB and their protein products.

• RNA viruses often carry oncogenes in their genomes.


Burkkit lymphoma
• The lymphoma is caused by Epstein-Barr virus through 3 different progression events:

The first step is infection with EBV which specifically infects B lymphocytes.

 The B cells are now immortalized, that is, they can be cultured indefinitely in artificial

medium.

 But they are still dependent on the B cell growth factor (BCGF) for proliferation.

 The second step is the chromosome translocation, usually from chromosome 8 to 14,

but sometimes from 8 to 2 or from 8 to 22.

 The chromosome 14 contains gene for immunoglobulin heavy chain, 2 contains gene for

kappa light chain and 22 for lambda light chain.


Cont’d...
 The transposing region in chromosome 8 contains the oncogene cmyc. This
transposition of oncogenes to one of the Ig Ioci confers BCGF-independence; but these B
cells can grow very slowly.

The third step is the activation of c-myc oncogene, with consequent malignancy. EBV is
also strongly associated with Nasopharyngeal carcinoma.

Similarly, in chronic myeloid leukemia, deletion of short arm of chromosome 22, called
Philadelphia (Ph') chromosome is seen in 80% cases. In the rest, there is translocation
of 9 to 22 leading to activation of c-abl present in chromosome 9.

In Non-Hodgkin's lymphoma, translocation of chromosome 14 to 18 is very common,


involving the bcl-2 oncogene. The bcl-2 product suppresses programmed cell death
leading to tumor formation.
Fig: Schematic representation of the reciprocal translocation involved in Burkitt lymphoma
HPV (Human Papilloma Virus)
HPV (Human Papilloma Virus) has a circular double stranded DNA. More than 100 HPV types
are known.

HPV types 16 and 18 are associated with human uterine cervical cancer; they cause 70% of all
cervical cancers HPV infects epithelial cells in the cervical mucosa; the virus multiplies and
lyses the host cells, causing a lesion.

In 99% of such cases healing occurs within 6 months to 2 years. But in about 1% cases, the HPV
DNA is integrated into some of the host cells. After about 10-30 years, these cells develop into
invasive cancer.

Vaccines against high risk HPV16 and 18 types are now developed that provide 95% protection

from infection of HPV, thereby reducing the chances of developing cervical cancer.
Hallmarks of cancer cells

• A neoplasm refers to any abnormal new growth of tissue. It may be benign or malignant in
nature. The term “cancer” is usually associated with malignant tumors.

• Tumors can arise in any organ in the body and result in different clinical features, depending on
the location of the growth.

Key properties of cancer cells:- they

(1) proliferate rapidly

(2) display diminished growth control

(3) Display increased genomic mutations at the level of the nucleotide, small and large insertions
and deletions (indels), and gross chromosomal rearrangements, duplications and loss;

(4) display loss of contact inhibition in culture in vitro;


Hallmarks of cancer cells
(5) invade local tissues and spread, or metastasize, to other parts of the body

(6) are self-sufficient in growth signals and are insensitive to antigrowth signals;

(7) stimulate local angiogenesis; and

(8) are often able to evade apoptosis.

These properties are characteristic of cells of malignant tumors. It is metastasis


that is generally responsible for the deaths of patients who have cancer.

By contrast, cells of benign tumors also show diminished control of growth, but do
not invade local tissue or spread to other parts of the body.
Fig: Major biologic features of cancer cells
Fig: Some biochemical and genetic changes that occur in human cancer cells
Relationship between cell
division, apoptosis and
cancer
Apoptosis
Many cells can precisely control the time of their own death by the process of
programmed cell death, or apoptosis (app’-a-toe’-sis; from the Greek for “dropping
off,” as in leaves dropping in the fall).

In the human body about 100,000 cells are produced every second by mitosis and a
similar number die by apoptosis!

Apoptosis is a peculiar well controlled individual cell death that is caspase mediated
and leads to fragmentation of the cell and organelles into numerous small buds, which
are then engulfed by macrophages without surrounding inflammation.
Importance of Apoptosis
1) Crucial for embryonic development

-Errors in Apoptosis can lead to Birth Defects

2) Important for maintaining homeostasis

- Cell death is balanced with mitosis to regulate cell number.

3) The monthly sloughing of cells of the uterine wall (menstruation) is another case of

apoptosis mediating normal cell death

3) Improper regulation contributes to human disease

- Neurodegenerative diseases: Parkinson’s and Alzheimer’s

- Cancer - Autoimmune diseases e.g. (diabetes type I)

- Viral diseases
Characteristics of apoptosis
It is a process that occurs in almost all living creatures since their early stages of
embryological development.

It is an active cytological process in which energy is consumed (ATP dependent)

It is programmed or controlled by genetic protocol or program (control of


enzymes, cell membrane surface proteins & cytoplasmic molecules, signal
transduction, gene expression)

It may be triggered by intrinsic or extrinsic stimuli


Morphology of Apoptosis
Cell shrinkage (condensation of cytoplasm)

Breakdown of mitochondria; release of cytochrome C

Nuclear condensation

Nuclear fragmentation

Cell membrane blebbing

Fragmentation; apoptotic body formation: membrane-bound cellular fragments,


which often lack nuclei

Phagocytosis
Cellular changes associated with apoptosis
Apoptosis cont’d…

Sometimes cell suicide is not programmed but occurs in response to biological


circumstances that threaten the rest of the organism.

For example, a virus-infected cell that dies before completion of the infection cycle
prevents spread of the virus to nearby cells.

Severe stresses such as heat, hyperosmolarity, UV light, and gamma irradiation also
trigger cell suicide; presumably the organism is better off with any aberrant,
potentially mutated cells dead.
Apoptosis cont’d…
There are three types of cell death.

1. Necrosis: It is also termed as cell murder. Cells undergo necrotic death if cell membrane
is damaged or due to decreased oxygen supply and if energy (ATP) production is blocked.

2. Apoptosis: occurs in tissue turnover. Individual cells or groups of cells undergo this type
of death. Aged cells in the body are removed by apoptosis. It is a genetically programmed
cell death. In the initial stages of apoptosis, cell shrinks, followed by fragmentation and
finally these fragments are eliminated by phagocytosis

3. Atrophy: This type of cell death occurs in the absence of essential survival factors.
Survival factors required by the cell are produced by other cells. Absence of nerve growth
factor leads to atrophy of nerves. It is also genetically programmed cell death
How Apoptosis Differs from Necrosis?
1. Apoptosis is intrinsically controlled, necrosis is not
2. Apoptosis is more rapid (12-24 hours) than necrosis
3. Apoptosis is induced by endogenous or exogenous stimuli, necrosis is always induced
by exogenous harms
4. Apoptosis is limited to single or few cells at a time, and occurs among healthy cell
population, necrosis is usually more extensive & occurs in tissue exposed to injuries
5. Cell cytoplasm shrinks in apoptosis and swells in necrosis.
6. Nucleosomes of apoptotic cells are 180 bp fragments, contrary to the irregular ones in
necrosis
7. Apoptosis has no inflammation, while necrosis leads to liberation of pro-inflammatory
mediators
8. Apoptosis has no systemic manifestations contrary to most inflammations
Mechanism of apoptosis
Four stages of apoptosis have been defined:

i. Commitment to death by extracellular or intracellular triggers/signals

ii. Cell killing (execution) by activation of intracellular proteases (caspases)

iii. Engulfment of cell corpse by other cells

iv. Degradation of the cell corpse within the lysosomes of phagocytic cells

Stimuli for Apoptotic Cell Death in Mammals

i. Growth factor deficiencies

ii. Ionizing radiation/viral infection

iii. Free radical toxicity

iv. Death receptor activation (such as Fas or CD95 triggering)


Fig: Stimuli for Apoptotic Cell Death
Death Factors

Definition: cytokines that activate an apoptosis program by binding to their specific


receptor. Typical examples of death factors are:

1. Fas ligand,

2. TNF (tumor necrosis factor) and

3. TRAIL (TNF-related apoptosis-inducing ligand).

- Apoptosis can also be induced by cytotoxic T-lymphocytes using the enzyme granzyme.

III. Activation of Caspase cascade

i. Various stimuli eventually activate the executioner (caspase) cascade

ii. Caspase cascades are apparently required for complete execution


Caspases are central initiators and executioners of apoptosis

All Caspases are synthesized as inactive proenzymes and all have a critical Cys residue at the active

site, and all hydrolyze their target proteins on the carboxyl-terminal side of specific Asp residues

(hence the name caspase, from Cys and Asp).

At least 14 different caspases exist in human cells

- Initiator caspases include: 2, 8, 9, 10 - Execution caspases include: 3, 6, 7

The caspase cascade can be activated by:

• Granzyme B released by cytotoxic T lymphocytes which is known to activate caspase-3 and -7;

• Death receptors (like FAS, TRAIL receptors and TNF receptor) which can activate caspase-8

and -10; and

• The apoptosome, regulated by cytochrome c and the Bcl-2 family, which activates caspase-9.
Fig: The extrinsic (death receptor-initiated) pathway of apoptosis
Cell injury e.g.,
radiation , toxins,
free radicals

Cytoplasmic bleb Apoptotic body


The intrinsic (mitochondrial) pathway of apoptosis. Death agonists cause changes in the inner mitochondrial membrane,
resulting in the mitochondrial permeability transition (MPT) and release of cytochrome c and other pro-apoptotic
proteins into the cytosol, which activate caspases. AIF= Apoptosis inhibitory factor; IAPs= Inhibitors of apoptosis
proteins; Apaf-1= apoptosis protease activating factor.
Intrinsic apoptosis pathway

Intrinsic apoptosis pathway involve procaspase-9 which is activated downstream of mitochondrial proapoptotic events at
the so called apoptosome, a cytosolic death signalling protein complex that is formed upon release of cytochrome c from
the mitochondria [Salvesen, 2002b]. In this case it is the dimerization of procaspase-9 molecules at the Apaf-1 scaffold that
is responsible for caspase-9 activation [Denault, 2002]. Once the initiator caspases have been activated, they can
proteolytically activate the effector procaspases-3, -6, and -7 which subsequently cleave a specific set of protein substrates,
including procaspases themselves, resulting in the mediation and amplification of the death signal and eventually in the
execution of cell death with all the morphological and biochemical features usually observed [Earnshaw, 1999].
BCL-2

 Bcl2 was the first apoptosis-related gene that was recognized to play a role in
tumorigenesis, and indeed, Bcl-2 is overexpressed in a variety of cancers, contributing
to cancer cell survival through direct inhibition of apoptosis.

 BCL-2 is a human proto-oncogene located on chromosome 18.

 Its product is an integral membrane protein (called Bcl-2) located in the membranes of
the endoplasmic reticulum (ER), nuclear envelope, and in the outer membrane of the
mitochondria.

 The gene was discovered as the translocated locus in a B-cell leukemia (hence the
name). This translocation is also found in some B-cell lymphomas.
Oncogens and oncogenes
Oncogenes
Oncogenes were originally discovered in tumor causing viruses(A definite proof for an
oncogene was first demonstrated in Rous sarcoma virus). The full virus produces
sarcoma in avians but a strain of virus deficient in a particular gene, could not cause
the disease. Hence this gene was named as sarcoma gene, abbreviated as src.

 Then later found to be derived from genes in the animal host cells, proto-oncogenes,
which encode growth-regulating proteins (The same DNA sequences are available in
normal avian cells also. This reveals that normal cells do contain DNA sequences
similar to viral oncogenes).

 To distinguish these two genes, they are denoted as Vsrc (viral gene) and C-src
(cellular gene).
Oncogenes cont’d…
Today, more than 100 human proto-oncogenes are known. They are located on specific
chromosomes.

Proto-oncogenes are important regulatory genes of the cells. In fact, viruses carry
these genes accidentally picking them from the host cells.

 During a viral infection, the host DNA sequence of a proto-oncogene is sometimes copied
into the viral genome, where it proliferates with the virus.

In subsequent viral infection cycles, the proto-oncogenes can become defective by
truncation or mutation.

Viruses, unlike animal cells, do not have effective mechanisms for correcting mistakes
during DNA replication, so they accumulate mutations rapidly.
Oncogenes cont’d…
When a virus carrying an oncogene infects a new host cell, the viral DNA (and
oncogene) can be incorporated into the host cell’s DNA, where it can now interfere
with the regulation of cell division in the host cell.

In an alternative, nonviral mechanism, a single cell in a tissue exposed to carcinogens


may suffer DNA damage that renders one of its regulatory proteins defective, with the
same effect as the oncogenic mechanism: failed regulation of cell division.

 Oncogenes are generated by “activation” of normal cellular proto-oncogenes, which


encode growth-stimulating proteins.
Oncogenes cont’d…
 The mutations that produce oncogenes are genetically dominant; if either of a pair of chromosomes contains
a defective gene, that gene product sends the signal “divide” and a tumor may result.

 The oncogenic defect can be in any of the proteins involved in communicating the “divide” signal.

 Oncogenes discovered thus far include those that encode:-

A. Secreted proteins

B. Growth factors: e.g. sis gene produces platelet derived growth factor (PDGF)

- Many of the oncogenes act through the production of growth factors. The growth factors generally cause
mitosis or differentiation of target cells.

- can act in an endocrine, paracrine, or autocrine manner and affect a wide variety of cells to produce a
mitogenic response.

- These may be considered as local hormones. There are more than 100 growth factors.

- Interleukins and interferons are growth factors released by lymphocytes/macrophages


Oncogenes cont’d…

C. Transmembrane proteins (receptors): e.g. erb-B produces receptor for EGF (epithelial
growth factor)

D. Cytoplasmic proteins (G proteins and protein kinases), e.g., src product, a membrane-
bound enzyme, phosphorylates a specific tyrosine residue, leading to cascade activation of
cellular events. Receptors for EGF, insulin, PDGF, etc. are also activated by src-product protein
and

E. The nuclear transcription factors that control the expression of genes essential for cell
division (Jun, Fos).

Some oncogenes encode surface receptors with defective or missing signal-binding sites,
such that their intrinsic Tyr kinase activity is unregulated.
Oncogenes cont’d…
For example, the oncoprotein ErbB is essentially identical to the normal receptor for
epidermal growth factor, except that ErbB lacks the amino-terminal domain that
normally binds EGF and as a result sends the “divide” signal whether EGF is present or
not.
Mutations in erbB2, the gene for a receptor Tyr kinase related to ErbB, are commonly
associated with cancers of the glandular epithelium in breast, stomach, and ovary.

Fig: Oncogene-encoded
defective EGF receptor
Oncogenes cont’d…

Mutant forms of the G protein Ras are common in tumor cells.

The ras oncogene encodes a protein with normal GTP binding but no GTPase activity.

The mutant Ras protein is therefore always in its activated (GTP-bound) form,
regardless of the signals arriving through normal receptors. The result can be
unregulated growth.

Mutations in ras are associated with 30% to 50% of lung and colon carcinomas and
more than 90% of pancreatic carcinomas.
Tumor suppresssor genes/Anti-oncogenes or Oncosuppressor Genes/
 A tumor suppressor gene produces a protein that normally suppresses
cell growth or cell division.
 When such a gene is altered by mutation, the inhibitory effect of its product is lost or
diminished. This loss-of-function of a tumor suppressor gene leads to increased cell growth
or division.
 As first suggested by AG Knudson in1971, based on studies of the inheritance
of retinoblastomas, both copies of a tumor suppressor gene must be affected for it to lose its
inhibitory effects on growth (ie, a mutated loss-of function allele, rb-, is recessive to a wild-
type RB copy of the gene).
 Retinoblastoma occurs in children and causes blindness if not surgically treated
 Very young children who develop retinoblastoma commonly have multiple tumors in both
eyes
 People with retinoblastoma who survive childhood also have a high incidence of cancers of
the lung, prostate, and breast later in life
Table: Important oncosuppressor genes
Gene of P53
 A part of short arm of chromosome 17 was shown to be deleted in various human cancers. This

region is now known to contain an oncosuppressor gene, called p53.

 Arnold Levine in 1979 identified the p53. It is so called because the gene encodes a phosphoprotein

with molecular weight 53,000; with 375 amino acids in length.

 It blocks the cells that have damaged DNA by triggering the production of another protein p21,

which blocks cell division until the damage is repaired.

 If the DNA damage is severe, p53 directs the cell to commit suicide by apoptosis program. It can

complex with other transforming proteins generated by other oncogenic viruses. (e.g. T antigen of

SV 40; E6 of HPV-16).

 Most tumors have a complete absence of p53, whereas others show mutant nonfunctional p53.
Properties of Some Important Oncogenes and Tumor Suppressor Genes
Table: Differences Between Oncogenes and Tumor Suppressor Genes
Stability genes(called caretaker genes)

Encode proteins that function in the repair of major genetic defects that result from

aberrant DNA replication, ionizing radiation, or environmental carcinogens.

Mutations in these genes lead to a high frequency of unrepaired damage (mutations) in other

genes, including proto-oncogenes and tumor suppressor genes, and thus to cancer.

Among the stability genes are:-

I. ATM ; the XP gene family, in which mutations lead to xeroderma pigmentosum

II. the BRCA1 genes associated with some types of breast cancer.
Summary
Three classes of defects can contribute to the development of cancer:

1. oncogenes, in which the defect is the equivalent of a car’s accelerator pedal being
stuck down, with the engine racing;

2. mutated tumor suppressor genes, in which the defect leads to the equivalent of
brake failure; and

3. mutated stability genes, with the defect leading to unrepaired damage to the cell’s
replication machinery, the equivalent of an unskilled car mechanic
Stages of carcinogenesis
Mutations in oncogenes and tumor suppressor genes do not have an all-or-none effect.

In some cancers, perhaps in all, the progression from a normal cell to a malignant tumor

requires an accumulation of mutations (sometimes over several decades), none of which, alone,

is responsible for the end effect.

For example, the development of colorectal cancer has several recognizable stages, each

associated with a mutation.

Stage 1: Benign polyp (early adenoma)

 If an epithelial cell in the colon undergoes mutation of both copies of the tumor suppressor

gene APC (adenomatous polyposis coli), it begins to divide faster than normal and produces a

clone of itself, a benign polyp (early adenoma)


Stages of carcinogenesis cont’d…
Stage 2: Intermediate adenoma

For reasons not yet known, the APC mutation results in chromosomal instability, and whole regions
of a chromosome are lost or rearranged during cell division. This instability can lead to another
mutation, commonly in ras, that converts the clone into an intermediate adenoma.

Stage 3: Late adenoma

A third mutation (often in the tumor suppressor gene DCC) leads to a late adenoma.

Only when both copies of p53 become defective does this cell mass become a carcinoma - a
malignant, life threatening tumor.

The full sequence therefore requires at least seven genetic “hits”: two on each of three tumor
suppressor genes (APC, DCC, and p53) and one on the proto-oncogene ras.
Fig: Multistep genetic changes associated with the development of colorectal cancers
Fig: Multistep transition from normal epithelial cell to colorectal cancer
Tumor markers (also called as tumor index substances)
 Cancer (tumour or malignant) cells produce abnormal substances.

 Usually these substances are not produced by normal cells.

 The abnormal substances produced by the cancer cells are enzymes, hormones and
proteins.

 These substances are released into blood by cancer cells. As a result their level in
blood rises.

 Measurement of these substances in blood or serum provides useful information


about cancer. Hence, they are called as tumour markers.

 Nowadays measurement of tumour markers in blood is an integral part of oncology.


Tumor markers cont’d…

 Tumour marker measurement is used in:-

(a) Detection of cancer.


(b) Diagnosis of cancer.
(c) Prognosis of cancer.
(d) Determination of cancer stage.
(e) Determination of location of cancer in the body.
(f) Determination of organ involved in cancer.
(g) Cancer therapy
Table: Common tumor
markers
Clinically Important Tumor Markers
1. Alpha Fetoprotein (AFP)

 It is 70,000 D molecular weight protein demonstrated by Abelev in 1963 AFP.

 The gene for AFP is located in chromosome No. 4

 It is fetal albumin and has similarities with adult albumin.

 It is increased in the circulation of patients with hepatocellular carcinoma, germ cell tumors,

teratocarcinoma of ovary and in pregnancy with fetal malformations of neural tube.

 In adult males and nonpregnant females, normal value is less than 15 ng/L.

 A value of AFP above 300 ng/L is often associated with cancer, although levels in this range may

be seen in nonmalignant liver diseases.

 Levels above 1000 ng/L are almost always associated with cancer (except in pregnancy).
2. Carcinoembryonic Antigen (CEA)

CEA level is markedly increased in colorectal cancers. Its molecular weight is 1,85 kD.

In 1965, Gold and Freedman identified the CEA.

 Over 50% of persons with breast, colon, lung, gastric, ovarian, pancreatic, and uterine
cancer have elevated levels of CEA.

CEA levels may also be elevated in inflammatory bowel disease (IBD), pancreatitis, and
liver disease.

Heavy smokers and about 5% of healthy persons have elevated plasma levels of CEA
3. Beta Chain of Chorionic Gonadotropin

Beta-HCG is synthesised by normal syncytia trophoblasts (cells of placental villi).

Its molecular weight is 45 kD. HCG is a glycoprotein; it has alpha and beta subunits.

The alpha subunit is identical with those of FSH, TSH and LH. The beta subunit is

specific for HCG.

It is increased in hydatidiform mole, choriocarcinoma and germ cell tumors.

About 60% of testicular cancers secrete hCG.

Normal value is less than 20 IU/L for males and non-pregnant females. Greater than

100,00 IU/ L indicates trophoblastic tumor.


4. Cancer Antigen 125 (CA-125)

CA-125 is a tumor maker for ovarian cancers.

It is a glycoprotein with a molecular weight of 10 million; one of the biggest molecules identified.

The name is so given because it reacted with a monoclonal antibody, originally termed as OC-125
(Bast, 1981).

Approximately 75% of persons with ovarian cancer will have elevated serum levels. (50% of
persons with stage I disease and 90% with stage II).

Elevated levels of CA-125 are also found in approximately 20% of persons with pancreatic and
digestive tract cancers. CA-125 levels correlate with tumor mass; consequently, this test is used to
determine whether recurrence of the cancer has occurred following chemotherapy.

Normal blood level of CA125 is less than 35 U/mL


5. Tissue Polypeptide Antigen (TPA)

It was isolated by Bjorklund in 1957.

It is a common human carcinoma antigen, produced during G2 phase and released
into surrounding fluids during mitosis.

It is not specific for cancer of a particular site; but it is useful to assess the activity of
the tumor.

It is seen in blood as long as the tumor cells proliferate.

The TPA blood test is sometimes used along with other tumor markers to help follow
patients being treated for lung, bladder, and many other cancers.
6. Prostate Specific Antigen (PSA)

It is a 32 kD glycoprotein isolated by Chu in 1980.

It is produced by secretory epithelium of prostate gland. It is normally secreted into seminal

fluid, where it is necessary for the liquefaction of seminal coagulum.

It is a protease, and in serum it is seen complexed with alpha-1-antitrypsin.

The PSA level, especially the complexed form, is increased in prostate cancers.

PSA has been found to be elevated in 60-70% patients with cancer of the prostate. Most PSA is

bound to antitrypsins in plasma but some PSA circulates unbound to protein (free PSA).

Normal blood level of total PSA is less than 4 ng/L. Persons with a borderline total PSA

(between 4-10 ng/L), but who have a low free PSA are more likely to have malignant prostate
Treatment of cancer
1. Chemotherapy
Compounds that block replication of cells and anti metabolites that block nucleotide
biosynthesis are used as anticancer agents or in chemotherapy of cancer.
Treatment of cancer
2. Radiotherapy

Radiation can break phosphodiester linkages of DNA and interferes with replication
process. As a result growth of cancer cells can come down. Based on this principle
radiation is used to treat tumours.
3. Surgery

 It is the treatment of choice in the advanced stages of cancer. Cancer (tumour) tissue
is removed by surgery. Usually surgery is performed with operataing microscope.
4. Photo chemotherapy

It is a newly introduced treatment for cancer. It uses a photosensitive drug and laser
light to destroy cancer cells.
Treatment of cancer
5. Suicide Gene Therapy (Molecular surgery)

It is a kind of gene therapy used in the treatment of solid tumours where therapeutic gene is
targeted at tumour cells killing cells which expressing it.

It is also known as molecular surgery. The suicide genes are enzymes which activates low toxic
prodrug to toxic potent drug.

Herpes simplex thymidine kinase (HSTK) and cytosine deaminase (CD) are two such
enzymes.

HSTK converts non-toxic anti-viral drug ganciclovir to toxic form by phosphorylation CD


converts non-toxic fluorocytosine into toxic fluorouracil.

Vectors carrying genes of these enzymes are injected directly into tumour. It is followed by
intratumoural injection of prodrug

You might also like