Monday, September 7, 2020

Cancer - Development and Causes of Cancer, Oncogenes and Tumor Suppressor genes, Tumor Viruses, Tumor Antigens, Immune responses to tumors and Cancer treatment

Cancer

Cancer is the abnormal growth of cells caused by multiple changes in gene expression.  These changes cause an imbalance of cell proliferation and cell death and the cells become able to invade tissues and metastasize to distant sites.  It is a genetic disease, but in most cases, not an inherited disease. 

Cancer is a complex family of diseases. Carcinogenesis is the events that turn a normal cell in the body into a cancer cell and is a complex multistep process.  Cancer is a disease of abnormal gene expression which happens by a number of mechanisms. These may be via a direct insult to DNA, such as a gene mutation, translocation, amplification, deletion, loss of heterozygosity, or from abnormal gene transcription or translation.

Tumors are of two basic types: benign and malignant. A tumor that is not capable of indefinite growth and does not invade the healthy surrounding tissue extensively is benign. A tumor that continues to grow and becomes progressively invasive is malignant.  Malignant tumors exhibit metastasis where small clusters of cancerous cells dislodge from a tumor, invade the blood or lymphatic vessels, and are carried to other tissues, where they continue to proliferate. In this way a primary tumor at one site can give rise to a secondary tumor at another site.

The following are features that differentiate a malignant tumor from a benign tumor:

1        Malignant tumors invade and destroy adjacent normal tissue.  Benign tumors grow by expansion, are usually encapsulated, and do not invade surrounding tissue.

2        Malignant tumors metastasize through lymphatic channels or blood vessels to lymph nodes and other tissues in the body. Benign tumors remain localized and do not metastasize.

3        Malignant tumor cells tend to be anaplastic (less differentiated than normal cells of the tissue in which they arise). Benign tumors usually resemble normal tissue.

4        Malignant tumors usually grow more rapidly than benign tumors. Benign tumors often grow slowly over several years.

The biological properties of cancer cells

The biological properties of malignant tumor cells are

  1. Acquisition of self-sufficiency in growth signals, leading to unchecked growth.
  2. Loss of sensitivity to anti-growth signals, leading to unchecked growth.
  3. Loss of capacity for apoptosis, that allows growth despite genetic errors and external anti-growth signals.
  4. The cells are attachment independent and has lost contact inhibition
  5. Loss of capacity for senescence, leading to limitless replicative potential (immortality)
  6. Acquisition of sustained angiogenesis, allowing the tumor to grow beyond the limitations of passive nutrient diffusion.
  7. Acquisition of ability to invade neighboring tissues, becoming invasive and acquisition of ability to build metastases at distant sites.

Carcinogenesis

Carcinogenesis is a general term used to denote the development of cancer. A carcinogen is any chemical, biological agent or radiation that has the ability to damage the genome or to disrupt cellular metabolic processes leading to cancer. 

Development of Cancer Occurs in Stages

Cancers are multistage diseases progressing through accumulation of multiple genetic changes (lesions) that influence control of cell proliferation, survival, differentiation, migration, and social interactions with neighboring cells and stroma.  In experimental models, the process has at least three distinct steps: initiation, promotion, and progression.

Initiation

Initiation can occur after a single, brief exposure to a carcinogen. Initiationinvolves changes in the genome.

Promotion

After initiation, promoters stimulate cell division and lead to malignant transformation.  Most promoting agents are mitogens for the tissue in which promotion occurs. Here the initiated cell clonally expands into a visible tumor, often a benign lesion such as a papilloma. 

Progression

The cells after promotion undergo one or more additional heritable changes during the progression to a malignant neoplasm.  During this loss of growth control and an escape from host defense mechanisms become predominant and progress to a clinically detectable tumor.

Spread of cancer from the organ of origin (primary site) to distant tissues (metastasis) occurs.

Tumor Metastasis

Tumor metastasis is the spread of tumor cells from a primary tumor to colonize other sites of the body. The formation of metastases is a very complex and dynamic process during which a number of interactions between tumor cells themselves and between tumor cells and the surrounding environment take place. The steps involved in the process of metastasis are the "metastatic cascade".

The tumor metastatic process is a complex cascade of events, including

1) Invasion, often defined by changes in tumor cell adhesion, proteinase production, and locomotion,

2) Intravasation, arrest in, and extravasation from the circulatory system,

3) Colonization,

4) Angiogenesis at a distant site.

At each step of the metastatic process tumor cells must avoid immune recognition and lysis.


Metastatic Cascade

Invasion and motility.

Normal tissue requires proper adhesions with basement membrane and/or neighboring cells to signal to each other. Tumor cells display diminished cellular adhesion, allowing them to become motile. Tumor cells use their migratory and invasive properties to burrow through surrounding extracellular stroma and to gain entry into blood vessels and lymphatics.

Intravasation and survival in the circulation

Once tumor cells enter the circulation, or intravasate, they must be able to withstand the physical shear forces and the hostility of immune cells to travel to distinct sites to establish the secondary tumor growth.  In the bloodstream, a very small number of tumor cells survive to reach the target organ.  NK cells, macrophages and lymphocytes contribute to the elimination of tumor cells in the vascular system. The death of circulating cancer cells may be also caused by very simple factors like mechanical movement, turbulence and lack of proper nutrition and metabolism.

Arrest and extravasation.

Once arrested in the capillary system of distant organs, tumor cells must extravasate, or exit the circulation, into tissue.

Growth in distant organs.

Successful adaptation of the extravasated cells to the new microenvironment results in their growth and development into metastatic tumour.

Pathologically, cancers are classified as:

1. Carcinoma, originating from epithelial cells in the skin or in tissues that line or cover internal organs, and typically representing over 80% of diagnosed human cancer each year;

2. Sarcoma, originating in bone, cartilage, fat, muscle, blood vessels, or other connective or supportive tissue;

3. Leukemia, a cancer originating in blood-forming tissues such as the bone marrow, and causing large numbers of abnormal blood cells to be produced and enter the bloodstream 

4. Lymphoma, which originates in the cells of the immune system.

Cancer involves changes in two classes of genes – onco genes and tumour suppressor genes

Oncogenes – stimulate proliferations, inhibit differentiation, inhibit apoptosis

Tumor suppressor genes- inhibit proliferation, promote differentiation, stimulate apoptosis

Gain-of-function mutations of proto-oncogenes stimulate cells to multiply.

Loss-of-function mutations of tumor suppressor genes cause uncontrolled multiplication.

Oncogenes (cancer-causing genes)

Proto-oncogenes are a family of cellular genes with at least 30 members that appear to be involved in normal cellular growth and development and play key roles in loss of differentiation, cell motility and avoidance of apoptosis.   Activation or inappropriate expression of these genes results in proliferative signals involved in neoplastic growth. In healthy cells, the proto-oncogenes are under tight regulation to avoid excessive proliferation. Its hyperactivity make it an oncogenes.

Oncogenes may be divided into five groups based on the functional and biochemical properties of protein products of their normal proto-oncogene counterparts. These groups are

(1) Growth factors,

(2) Growth factor receptors,

(3) Signal transducers,

(4) Transcription factors,

(5) Regulators of cell death and others.

Conversion of proto-oncogenes to oncogenes. A proto-oncogene can be converted to an oncogene in a number of ways.

1.      Point mutations in the coding region of a single gene (e.g. RAS) that can result in the formation of an abnormal oncoprotein with enhanced stability or activity

2.      Point mutations in regulatory elements resulting in enhanced or deregulated expression.

3.      Chromosomal translocations or rearrangements can lead to over expression of an oncoprotein.

For example, in Burkitt’s lymphoma, the proto-oncogene c-MYC on chromosome 8 is translocated to one of the three chromosomes containing the genes that encode antibody molecules: immunoglobulin heavy chain locus (chromosome 14) or one of the light chain loci (chromosome 2 or 22). C-MYC now finds itself in a region of vigorous gene transcription, leading to overproduction of the c-MYC protein.

The BCL-2 Family comprises both proapoptotic and antiapoptotic members, the balance determines whether or not a cell undergo apoptosis.

Bcl-2 subfamily (antiapoptotic): Bcl-2, Bcl-XL, Bcl-w, etc.

Bax subfamily (pro-apoptotic): Bax, Bak and Bok

BH3 subfamily (pro-apoptotic): Bad, Bid, Bik, etc.

BCL-2 in human follicular lymphoma involves a chromosome translocation event that moves the BCL-2 gene from chromosome 18 to 14 linking the BCL-2 gene to an immunoglobulin locus. The Bcl-2 gene implicated in a number of cancers - melanoma, breast, prostate and lung carcinomas and involved in resistance to conventional cancer treatment.

4.      Gene amplification can lead to overexpression of the oncogene.

5.      Fusion of one protein to another might lead to its constitutive activity.

Reciprocal translocation between the ABL chromosome (chromosome 9) and chromosome 22 near a locus termed the break-point cluster region (BCR). The result is a constitutively active ABL tyrosine kinase domain fused to the BCR coding region forming the BCR-ABL fusion protein - the Philadelphia chromosome (Ph+) - chronic myelogenous leukemias (CMLs).

Fusion of the promyelocytic leukemia (PML) protein to the retinoic acid receptor-alpha (RARα) generates the transforming protein of acute promyelocytic  leukemias

Tumor suppressors

These are guardians against DNA damage that may be induced by ultraviolet (UV) exposure from sunlight, gamma irradiation (X-rays), chemotherapeutic drugs, or an excess of inappropriate proliferative signals. Tumor suppressor genes prevent cells from becoming malignant by arresting their proliferation or inducing them to commit suicide (apoptosis).

Tumor suppressors monitor critical cellular checkpoints that govern the mitotic cycle, DNA repair, transcription, apoptosis, and differentiation. The functional inactivation of tumor suppressors by mutation, deletion, or gene silencing creates an imbalance between proliferation, cell death, and differentiation programs that facilitates tumorigenesis. 

Example is p53/TP53 “The guardian of the genome” - a transcription factor - its loss leads to genomic instability and increased mutagenesis.

BRCA1 and BRCA2 are tumor suppressor genes and are involved in DNA repair of double-strand breaks.  Mutations in either the BRCA1 or BRCA2 genes - high risk for breast and ovarian cancer phenotypes,

Haploinsufficiency is defined by the appearance of a phenotype in cells or an organism when only one of the two gene copies or alleles is inactivated. For some tumor suppressor genes, the loss of a single allele is sufficient to induce susceptibility to tumor formation and these are haploinsufficient tumor-suppressor genes.

According to Knudson’s “two hit” hypothesis, loss of heterozygosity is to be achieved to develop retinoblastoma in the eyes of children. Here both copies of the gene must be inactivated for cancer to occur.

Carcinogens

Carcinogens are agents that may damage genes involved in cell proliferation and migration, and may selectively enhance growth of tumor cells or their precursors. Three classes of environmental agents – chemical carcinogens (cancer causing chemicals), ionizing radiation and viruses or other microorganisms (that cause cancer in animals) have been shown to increase the risk of cancer.  They induce mutations that lead to cancer.

The importance of mutagenesis in the induction of cancer could be exemplifyied by diseases such as xeroderma pigmentosum. This disorder is caused by a defect in the gene that encodes a DNA-repair enzyme called UV-specific endonuclease. Individuals with this disease are unable to repair UV-induced mutations and consequently develop skin cancers.

Carcinogenic chemicals

Carcinogenic chemicals such as azo dyes, asbestos, benzene, formaldehyde, and diesel exhaust are dangerous in high concentrations. Chemical carcinogens taken up by cells are usually metabolized, and the resulting metabolites are then excreted, but may on occasion be retained by the cell. Such internalized carcinogenic compounds can then directly or indirectly alter gene expression. Some carcinogens are genotoxic, forming DNA adducts or inducing various chromosomal abnormalities. For example, carcinogenic ions or compounds of nickel, arsenic, and cadmium can induce aneuploidy. Many carcinogenic compounds are mutagenic – that is, they can induce mutations in DNA.

Other carcinogens may act by nongenotoxic mechanisms, including promoting inflammation, suppressing immunity, forming damaging reactive oxygen species (ROS), or by activation of signaling pathways and epigenetic silencing. 

Some carcinogens are direct-acting and activation-independent.  Example is alkylating agents. However, the majority are procarcinogens that require metabolic activation to produce carcinogens. Polycyclic hydrocarbons (smoke), aromatic amines, amides, azo dyes, and nitrosamines require activation by the hepatic cytochrome P450 mixed function oxidase system.

The result of exposure of a cell to certain carcinogens permanently alters its genetic material but does not immediately influence phenotype and these are described as mutagens or genotoxic agents and these are initiators. Promoters are nongenotoxic carcinogens. The best-known promoters are the phorbol esters.

Ionizing radiation:

Radiation-induced cancers may represent up to 10% of total cases.  X-rays and nuclear radiation can damage DNA. Radiation-induced cancers are a stochastic late effect of ionizing or non-ionizing radiation. They include some leukemia and lymphoma, thyroid cancers, skin cancers, some sarcomas and some lung and breast carcinomas.

Viruses and other microorganisms:

It is estimated that oncogenic viruses are involved worldwide in about 16% of neoplasia. Few  DNA viruses and diploid RNA viruses have been shown to be associated with human cancers. 

DNA viruses

Cellular transformation by DNA tumor viruses is due to the result of protein–protein interaction. Proteins encoded by the DNA tumor viruses, the tumor antigens, or T antigens, can interact with cellular proteins mainly of the tumorsuppressor type. The loss of normal suppressor functions results in cellular transformation.

Hepadnaviruses Hepatitis B virus (HBV) causes hepatocellular carcinoma.

Papillomavirus - Human papilloma viruses (HPV) cause common warts and cervical carcinoma.

Herpes viruses - Epstein–Barr virus (EBV), the etiological agent of infectious mononucleosis cause Burkitt’s lymphoma and nasopharyngeal carcinoma, Hodgkin’s lymphomas. EBV transformation is multigenic and at least five different viral genes appear to be involved.

Adenoviruses - These viruses transform cultured cells and cause cancer in animals.

Polyomaviruses - Oncogenic in laboratory animals and can transform human cells in culture.  Polyoma virus and SV40 are linked to a variety of animal tumors. The latter is known to inactivate both the p53 and RB tumor suppressors.

Retroviruses

RNA tumor viruses in humans are Human T-cell leukemia viruses (HTLVs) and the related retrovirus, human immunodeficiency virus (HIV).

Tumorigenic retroviruses (oncoviruses) are grouped into three categories based on their mechanism of oncogenicity:

(i) transducing retroviruses;  (ii) cis-activating retroviruses  (iii) trans-activating retroviruses.

Two features of the retrovirus life cycle make them acquire and activate oncogenes.

Integration into the cell chromosome allows the virus to take control of and modify cellular genes.  Most retroviruses do not kill the cells they infect so that genetic alterations are transmitted to daughter cells.

When a retrovirus infects a cell, its RNA genome is converted into DNA by the viral encoded RNA-dependent DNA polymerase (reverse transcriptase). The DNA can then integrate stably into the genome of the host cell.  It will be copied as the host genome is duplicated during normal cell division.

Transducing retroviruses

Integration in cellular genome may result in rearrangement of the viral genome and sometimes a portion of the host genome will be incorporated into the viral genome.  This is termed transduction.

Occasionally, transduction results in the virus acquiring a host gene involved in cellular growth control.  The transduced gene is always altered by either point mutation or deletion. These alterations serve to activate the transduced gene and result in uncontrolled cellular proliferation. The transduced genes are termed oncogenes. The expression of the transduced gene is driven by the virus promoter/enhancer region (LTR). Long terminal repeats (LTRs) are powerful transcriptional promoter sequences found at the ends of the retroviral genome. Transducing retroviruses cause tumors at high efficiency (100% of animals) and with short latency periods (days).

Cis-activating retroviruses

The second mechanism for retroviral transformation of cells is through the transcription-promoting action of the LTRs. Retroviral genomes integrate randomly into the host genome, and may occasionally result in the viral LTRs being placed near to a gene that encodes a growth regulating protein. This may result in over expression of the growth-regulatory gene and result in cellular transformation. This is termed retroviral integration-induced transformation. Tumors formed by these viruses take longer to occur and not always form tumors.

Example is the induction of certain cancers by HIV infection.

Trans-acting retroviruses

These viruses may upregulate cell oncogenes through the action of a viral transactivator protein. The latency period is long (years) and the efficiency of tumor induction is very low (1%).

HTLV-1, Human T-cell leukemia virus is example.

In addition to viruses, some microflora bacteria of the gastrointestinal tract, including Helicobacter pylori for gastric neoplasia, some parasites such as Opisthorchis viverrini for gallbladder cancers, Schistosoma haematobium for bladder cancer and some oncogenic toxins from moulds inducing aflatoxins-related HCC are also identified.

Tumor Antigens

Tumor antigen is an antigenic substance produced in tumor cells, i.e., it triggers an immune response in the host. Tumor antigens are useful tumor markers in identifying tumor cells with diagnostic tests and are potential candidates for use in cancer therapy.

The use of a variety of genetic, biochemical, and immunological approaches has allowed the identification of several tumor-associated antigens.  Common tumor antigens offer hope for the design of better therapies, detection, and monitoring, and have important implications for the possibility of anti-tumor immunization.

There are 2 main types of tumor antigens

Tumor-specific transplantation antigens (TSTA) are unique to tumor cells and not expressed on normal cells. They may result from mutations in tumor cells that generate altered cellular proteins.  Processing of these proteins gives rise to novel peptides that are presented with class I MHC molecules and induce a cell-mediated response by tumor-specific CTLs. They are responsible for rejection of the tumor. 

Tumor associated transplantation antigens (TATA) that are expressed by tumor cells and normal cells. They may be expressed at higher levels on tumor cells when compared to normal cells. Alternatively, they may be expressed only during development of cells and lost during adult life but re-expressed in tumors.

These antigens may be proteins that are expressed on normal cells during fetal development but not expressed in the adult. They are termed Oncofetal tumor antigens. During embryonic development, the immune system is immature and unable to respond. Reactivation of the embryonic genes that encode these proteins results in their expression on tumor cells.  Two well-studied oncofetal antigens are alpha-fetoprotein (AFP) and carcinoembryonic antigen (CEA).

Tumor-associated antigens may also be proteins that are normally expressed at extremely low levels on normal cells but are expressed at much higher levels on tumor cells.  Examples are growth factors and growth-factor receptors and oncogene-encoded proteins.

For instance, a variety of tumor cells express the epidermal growth factor (EGF) receptor at levels 100 times greater than that in normal cells.

Tumor associated antigens encoded by cellular oncogenes are also present in normal cells where they are encoded by the corresponding proto-oncogene. But the levels will be higher on tumor cells.  For example human breast-cancer cells exhibit elevated expression of the oncogene-encoded Neu protein, a growthfactor receptor, whereas normal adult cells express only trace amounts of Neu protein.

Many tumor antigens are cellular proteins that give rise to peptides presented with MHC molecules.   These antigens are able to induce the proliferation of antigen-specific CTLs or helper T cells.  The tumor antigens recognized by human T cells fall into one of four major categories:

1.      Antigens encoded by genes exclusively expressed by tumors

2.      Antigens encoded by variant forms of normal genes that have been altered by mutation

3.      Antigens normally expressed only at certain stages of differentiation or only by certain differentiation lineages

4.      Antigens that are overexpressed in particular tumors

Chemically or physically induced tumor antigens

Even when the same chemical carcinogen induces two separate tumors at different sites in the same animal, the tumor antigens are distinct and the immune response to one tumor does not act against the other tumor. Methylcholanthrene and ultraviolet light are examples. 

Tumor Antigens May Be Induced by Viruses

In contrast to chemically induced tumors, virally induced tumors express tumor antigens shared by all tumors induced by the same virus.

Immune responses to tumors

The immune surveillance theory by Paul Ehrlich suggested that cancer cells frequently arising in the body are recognized as foreign and are eliminated by the immune system. Tumors arise only if cancer cells are able to escape immune surveillance, either by reducing the tumor antigen expression or due to any impairment in the immune system.

Cytotoxic factors such as TNF-α and TNF-β mediate tumor-cell killing. The immune response to tumors includes

·         CTL-mediated lysis

·         NK-cell activity

·         Macrophage-mediated tumor destruction

·         Cell destruction mediated by ADCC

NK cells and Macrophages play a significant role in the immune response to tumors and since they are not MHC restricted and they express Fc receptors, they can bind to antibody on tumor cells and mediate ADCC. activated macrophages secrete tumor necrosis factor α.

Tumors may evade the immune response by modulating their tumor antigens or by reducing their expression of class I MHC molecules or by antibody mediated or immune complex-mediated inhibition of CTL activity.

Cancer therapy

Conventional cancer treatment modalities are surgery, chemotherapy and radiation therapy.

Complete removal of the cancer without damaging the normal tissue of the body is the goal. Sometimes this can be accomplished by surgery, but this may increase the chance of cancer to invade adjacent tissue or to spread to distant sites by microscopic metastasis and chemotherapy and radiotherapy destroy normal cells too.  Therefore, cure may be associated with some level of adverse effects.  Cancer therapy can also include suppressing the cancer to a subclinical state and maintaining it subsided and palliative care where there is no hope for cure.

Several new methods for specific and targeted cancer treatment are now being emerging.  These are listed below.

Molecular approaches of cancer therapy

Targeted therapies

Immunotherapy

Hormone therapy

Angiogenesis inhibitors

Gene Therapy

Virotherapy

Targeted therapy is currently a very active research area. Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell. Examples are the tyrosine kinase inhibitors imatinib and gefitinib

Immunotherapy
Immunotherapy has been used as a novel means of treating cancer. Both active and passive means of stimulating the non-specific and specific immune systems have been employed.

Active Immunotherapy

In this, the host actively participates in mounting an immune response

Specific activation is achieved by using vaccines:

i) Hepatitis B vaccine

ii) Human Papilloma virus (HPV) vaccine

Nonspecific activation is achieved by immunization with:

i) Bacillus Calmette-Guerin (BCG)

ii) Corynebacterium parvum

These activate macrophages to be tumoricidal.

Passive Immunotherapy

This involves transfer of preformed antibodies, immune cells and other factors into the hosts.

Specific Passive Immunotherapy

1) Antibodies against tumor antigens (e.g. Her2/Neu for treatment of breast cancer)

2) Antibodies against IL-2R for Human T lymphotropic virus (HTLV-1)-induced adult T cell leukemia

3) Antibodies against CD20 expressed on non Hodgkin’s B cell lymphoma.

These antibodies bind to tumor antigens on the cell surface and activate complement to mediate tumor cell lysis. In addition cells such as NK cells, macrophages and granulocytes may bind to the antigen-antibody complexes on the tumor cell surface and mediate tumor cell killing through antibody-dependent cell-mediated cytotoxicity.

4) Antibodies conjugated to toxins, radioisotopes and anti-cancer drugs have also been used. These enter the cells and inhibit protein synthesis. e.g. anti-CD20 conjugated to Pseudomonas toxin or ricin toxin.

Anti-HER2/neu antibody trastuzumab (Herceptin) is used in breast cancer, and the anti-CD20 antibody rituximab is used in a variety of B-cell malignancies

The problems associated with the use of antibodies

      Antibodies are not efficient because the tumor antigens are associated with class I MHC antigens.

      Tumors may shed antigen or antigen-antoibody complexes. Thus, immune cells cannot mediate tumor destruction.

      Some antibodies may not be cytotoxic.

      Antibodies may bind non-specifically to immune cells expressing the Fc receptors which include NK cells, B cells, macrophages and granulocytes without binding to tumor cells.

Nonspecific Passive Immunotherapy

i) Adoptive Transfer of lymphocytes:  Lymphokine-activated killer (LAK) cells or Tumor-infiltrating lymphocytes (TIL)

ii) Dendritic cells activated with tumor antigens may induce tumor-specific T cell responses. As tumor Ags are usually not known, tumor lysates are used.

iii) Cytokines

§    IL-2: Activates T cells/NK cells expressing IL-2 receptors. This is used in the treatment of renal cell carcinoma and melanoma

§    IFN-alpha: This induces MHC expression on tumors and used in the treatment of hairy B cell leukemias

§    IFN-gamma: This increases class II MHC expression; used in the treatment of ovarian cancers.

§    TNF-alpha: This kills tumor cells.

iv) Cytokine gene transfected tumor cells may also be used which can activate T or LAK cell-mediated anti-tumor immunity.

Hormone therapy

The growth of some cancers can be inhibited by providing or blocking certain hormones. Examples of hormone-sensitive tumors include certain types of breast and prostate cancers. Removing or blocking estrogen or testosterone is done as treatment.

Angiogenesis inhibitors

Angiogenesis inhibitors prevent the extensive growth of blood vessels (angiogenesis) that tumors require to survive. Bevacizumab is an example.

Gene Therapy

Gene therapy involves an approach that aims to modify, delete, or replace abnormal gene or genes in a target cancer cell.  Several methods have been developed to facilitate the entry of genetic materials (transgenes) into target cells, using various vectors.

Once genetic materials are transferred into target cells and incorporated into nuclear genetic DNA, they may induce silencing, down-regulation, modification, or repair of the target cell genes. This may lead to cell death and tumor necrosis (as with the suicide gene), or impaired cell growth with tumor regression (as with the silencing gene). Modification of the gene may improve the response from subsequent cancer therapy, such as chemotherapy, immunotherapy, or radiation. 

Suicide genes are transgenes that make up products that can cause a cell to kill itself through apoptosis. 

Gene silencing has been achieved through delivery of a small interfering double-stranded RNA (siRNA) into target cells, and subsequent duplex formation of RNA-induced silencing complex (RISC) that destroys messenger-RNA (mRNA), thus leading to interference with RNA functions and protein synthesis within the target cells.

Gene modification is helpful in improving cancer therapy, such as with radiation therapy. Radiosensitizing gene therapy promotes transgene expression in tumor tissue, thus increasing tumor sensitivity to radiation.

Gene repair can be achieved using zinc finger nuclease attached to the lentiviral vector. Once the viral vector enters the nucleus, it binds to a specific location in the double-stranded DNA, breaking it at specific location, with subsequent endogenous repair mechanisms, to create a newly edited double-stranded DNA.

The vectors are broadly divided into two major categories: viral (or bacterial) and non-viral vectors.  Viruses usually bind to target cells and introduce their genetic materials and transgenes into the host cell as part of their replication process. For non-viral vectors, different approaches have been utilized, using physical, chemical, as well as other modes of genetic transfer such as Electroporation, nanoparticles, liposomes, etc.

Virotherapy - Newcastle disease virus, herpes simplex virus, Adenovirus, Vaccinia virus, etc are under research and clinical trials to be used as oncolytic virus for treatment of tumours


No comments:

Post a Comment