Oncoimaging: Basic science to clinical application



2.15: Oncoimaging: Basic science to clinical application


2.15.1

CANCER BIOLOGY


Prerit Sharma, Abhishek Mahajan, Shreya Shukla, Shivam Rastogi, Pritesh Shah



Introduction


Cancer is a multifactorial disease, and imaging forms an important platform to demonstrate various mechanisms and phases of pathogenesis. Combining multiple imaging modalities and biomarkers improves diagnostic sensitivity and specificity. As a result, biomedical imaging plays a significant role in cancer management. These include prediction, screening, biopsy guidance for detection, staging, prognosis, therapy planning, therapy guidance, therapy response, recurrence and palliation.


The basis of modern pathology is understanding the cellular and molecular aberrations that give rise to diseases. This chapter discusses basic principles and emphasizes recent advances relevant to cancer biology (Table 2.15.1.1).



TABLE 2.15.1.1


Growth Factors Involved in Regeneration and Repair







































Growth Factor Sources Functions
Epidermal growth factor (EGF) Activated macrophages, salivary glands, keratinocytes, many other cells Mitogenic for many cell types: stimulates epithelial cell migration: stimulates formation of granulation tissue
Transforming growth factor-a (TGF-α) Activated macrophages, keratinocytes, many other cells Stimulates proliferation of hepatocytes and many other epithelial cells
Hepatocyte growth factor (HGF) (scatter factor) Fibroblasts, stromal cells in the liver, endothelial cells Enhances proliferation of hepatocytes and other epithelial cells: increases cell motility
Vascular endothelial growth factor (VEGF) Mesenchymal cells Stimulates proliferation of endothelial cells: increases vascular permeability
Platelet-derived growth factor (PDGF) Platelets, macrophages, endothelial cells, smooth muscle cells, keratinocytes Chemotactic for neutrophils, macrophages, fibroblasts, and smooth muscle cells: activates and stimulates proliferation of fibroblasts, endothelial cells, and other cells: stimulates ECM protein synthesis
Fibroblast growth factors (FGFs) including acidic (FGF-I) and basic (FGF-2) Macrophages, mast cells, endothelial cells, many other cell types Chemotactic and mitogenic for fibroblasts; stimulates angiogenesis and ECM protein synthesis
Transforming growth factor-β (TGF-fJ) Platelets, T lymphocytes, macrophages, endothelial cells, epithelial cells, smooth muscle cells, fibroblasts Chemotactic for leukocytes and fibroblasts: stimulates ECM protein synthesis: suppresses acute inflammation
Keratinocyte growth factor (KGF) (i.e., FGF-7) Fibroblasts Stimulates keratinocyte migration, proliferation, and differentiation

ECM, Extracellular matrix.


Reproduced from Vinay Kumar, Abul Abbas, Jon Aster. Robbins Basic Pathology, 10e, The Cell as a Unit of Health and Disease, Table 1.1, Philadelphia, Elsevier Inc., 2018.


The genome


Approximately 3.2 billion D.N.A. base pairs constitute the human genome. Of this entire genome, there are only about 20,000 protein-encoding genes (1.5%). These serve as the blueprints that instruct the assembly of enzymes, structural elements, and signalling molecules for an approx. The number of 50 trillion cells together constitutes the physical body.


There are five major classes of available non–protein-coding sequences in the human genome (Fig. 2.15.1.1):




  • Promoter and enhancer regions – serve as binding sites for transcription factors.
  • Binding sites – for factors that organize and maintain higher-order chromatin structures.
  • Non-coding regulatory R.N.A.s: Over 60% of the genome is transcribed into R.N.A.s. These regulate organic phenomena through a spread of mechanisms and are never translated. There are two best-studied varieties – micro-RNAs (miRNAs) and long non-coding R.N.A.s (lncRNAs).
  • Mobile genetic elements (e.g. transposons) structure one-third of the human genome. Mobile genetic elements have a role in gene regulation and chromatin organization. These genes move around the genome at the time of evolution, resulting in variability in the numbers and positions of the copies even among closely related species of humans and other primates. Nevertheless, their function isn’t well established. There are unique structural regions of D.N.A., namely, telomeres (chromosome ends) and centromeres (chromosome ‘tethers’). Satellite D.N.A. makes a significant component of centromeres, consisting of large arrays of repeating sequences (from five bp up to 5 kb). Although classically related to spindle apparatus attachment, satellite D.N.A. has a vital role in maintaining heterochromatin’s dense, tightly packed organization. The two most typical types of D.N.A. variation within the human genome are single nucleotide polymorphisms (SNPs) and replica number variations (CNVs). It is essential to notice that alterations in the D.N.A. sequence cannot by themselves explain the range of phenotypes in human populations.
  • Histone organization: Histones are highly basic proteins. These are abundant in lysine and arginine residues found in eukaryotic cell nuclei. They act as spools around which D.N.A. entwines to create structural units called nucleosomes. These are then wrapped into 30-nanometer fibres that form tightly packed chromatin.
  • Micro-RNA and Long Non-coding R.N.A.: Genes also can be controlled by non-coding R.N.A.s. These sequences are transcribed but not translated. Micro-RNA: miRNAs don’t encode proteins; they modulate the translation of target messenger R.N.A.s (mRNAs). The total number of microRNA in human genome is around 6000 which make up about 30% of the protein-coding genes. One miRNA can be responsible for multiple protein-coding genes which allow the coregulation of each miRNA to the entire program of the organic phenomenon.
  • Long Non-coding R.N.A.: Recent studies have identified a whole new galaxy of lncRNAs – by some calculations, the number of lncRNAs may exceed coding mRNAs by 10-fold–20-fold. lncRNAs regulate gene expression by several mechanisms.
  • Gene Editing: Gene editing repurposes this process by using artificial 20-base guide R.N.A.s (gRNAs) that bind Cas9 and complement a targeted D.N.A. sequence.

Image
Fig. 2.15.1.1 The organization of nuclear DNA At the light microscopic level, the nuclear genetic material is organized into dispersed, transcriptionally active euchromatin and densely packed, transcriptionally inactive heterochromatin; chromatin can also be mechanically connected with the nuclear membrane, and membrane perturbation can thus influence transcription. Chromosomes (as shown) can be visualized only during mitosis. During mitosis, they are organized into paired chromatids connected at centromeres; the centromeres act as the locus for the formation of a kinetochore protein complex that regulates chromosome segregation at metaphase. The telomeres are repetitive nucleotide sequences that cap the termini of chromatids and permit repeated chromosomal replication without deterioration of genes near the ends. The chromatids are organized into short “P” (“petite”) and long “Q” (next letter in the alphabet) arms. The characteristic banding pattern of chromatids has been attributed to relative GC content (less GC content in bands relative to interbands) with genes tending to localize to interband regions. Individual chromatin fibers are comprised of a string of nudeosomes—DNA wounds around octameric histone cores—with the nudeosomes connected via DNA linkers. Promoters are noncoding regions of DNA that initiate gene transcription; they are on the same strand and upstream of their associated gene. Enhancers can modulate gene expression over distances of 100 kb or more by looping back onto promoters and recruiting additional factors that drive the expression of premessenger RNA (mRNA) species. Intronic sequences are spliced out of the pre-mRNA to produce the final message that is translated into protein—without the 3′-untranslated region (UTR) and 5′-UTR. In addition to the enhancer, promoter, and UTR sequences, noncoding elements, including short repeats, regulatory factor binding regions, noncoding regulatory RNAs, and transposons, are distributed throughout the genome. Source: (Reproduced from Vinay Kumar, Abul Abbas, Jon Aster. Robbins & Cotran Pathologic Basis of Disease, 10e, The Cell as a Unit of Health and Disease, Figure 1.1, Philadelphia (PA), Elsevier Inc., 2021.)

Cell structure and organelles (Fig. 2.15.1.2)


Eukaryotic cells are different from bacterial or archaeal cells as they contain a membrane-bound nucleus. In addition, highly organized physical structures called intracellular organelles such as the endoplasmic reticulum, Golgi apparatus, chloroplasts and mitochondria are found in eukaryotic cells. The physical and chemical constituents of the cell organelles hold great significance for normal cell functioning.


Image
Image
Fig. 2.15.1.2 Basic subcellular constituents of cells. The table presents the number of the various organelles within a typical hepatocyte as well as their volume within the cell. The figure shows geographic relationships but is not intended to be accurate to scale. ER, Endoplasmic reticulum.

Cell membrane


The cell membrane (a.k.a. plasma membrane) is a thin, pliable, elastic structure surrounding the eukaryotic cell. It is about 7.5–10 nm in thickness and is composed almost entirely of proteins and lipids. It regulates the passage of organic molecules, ions, water, and oxygen into and out of the cell. In addition, waste products (such as CO2 and ammonia) leave the cell by passing through the cell membrane, often requiring the assistance of protein transporters.


One of the essential functions of the plasma membrane is protection and nutrient acquisition. This function is accomplished with the help of membrane components such as phosphatidylinositol, phosphatidylserine, glycolipids and sphingomyelin.


The plasma membrane is engraved with a variety of proteins and glycoproteins, whose functions are namely:




  1. 1. Trans-membrane transport of ions and metabolites.
  2. 2. Fluid-phase and receptor-mediated uptake of macromolecules.
  3. 3. Various intercellular interactions as well as interactions between cell-ligand and cell-matrix.

Many proteins that constitute the plasma membrane function as large complexes. Most proteins are integral or transmembrane proteins. These have one or more hydrophobic α-helical segments that traverse the lipid bilayer. Post-translational modification can be done on proteins synthesized on free ribosomes within the cytosol. The methods include adding prenyl groups (e.g., farnesyl, associated with cholesterol) or fatty acids (e.g., palmitic or myristic acid) that insert into the cytosolic side of the cell wall. Post-translationally added glycosylphosphatidylinositol tails anchor proteins on the extracellular face of the membrane. Peripheral membrane proteins may bond with true transmembrane proteins by covalent bonds. The transport of molecules across the lipid bilayer or into the intracellular sites via vesicular transport occurs via several mechanisms such as (1) Passive Diffusion; (2) Carriers and Channels; and (3) Receptor-Mediated and Fluid-Phase Uptake.


Cytoskeleton


An intracellular scaffold of structural proteins from the cytoskeleton determines cells’ ability to adopt a specific shape, maintain polarity, organize intracellular organelles and migrate. The eukaryotic cells have three major classes of cytoskeletal proteins:




  • Actin microfilaments
  • Intermediate filaments
  • Microtubules

Cell–cell interactions


The junctional complexes form mechanical links between cells and facilitate receptor-ligand interactions. The cells connect and communicate via these junctional complexes. Similar junctional complexes also mediate interaction with the E.C.M. Functionally, the most significant of these junctions are




  • Occluding junctions (tight junctions)
  • Anchoring junctions (adherens junctions and desmosomes)
  • Communicating junctions (gap junctions)

Cytoplasm


Both minuscule and large dispersed particles and organelles are dispersed within the cytoplasm. Cytosol is the jelly-like component of the cytoplasm which is made up of dissolved electrolytes, glucose and proteins. Dispersed within the cytoplasm are neutral fat globules, glycogen containing granules, ribosomes, secretory vesicles and five vital organelles: the endoplasmic reticulum, the Golgi body, mitochondria, lysosomes and peroxisomes.



Biosynthetic Machinery: Endoplasmic Reticulum and Golgi Apparatus


In an ongoing process of balancing synthesis and degradation, all cellular constituents, including structural proteins, enzymes, transcription factors and even the phospholipid membranes, are constantly renewed.



Endoplasmic Reticulum (E.R.) is a network of flat, tubular, vesicular structures in the cytoplasm. This organelle aids in processing molecules produced by the cell and transports these molecules to their destinations inside or outside the cell. Their walls are composed of lipid bilayer membranes containing large proteins, like the cell wall. Ribosomes are minute granular particles attached to the endoplasmic reticulum’s outer surfaces. The reticulum is named the granular endoplasmic reticulum, where these particles are present. The ribosomes are composed of a mix of R.N.A. and proteins. Their primary function is to synthesize new protein molecules. Part of the endoplasmic reticulum that has no attached ribosomes is called the agranular or smooth endoplasmic reticulum. Smooth E.R. synthesizes lipid substances and is responsible for other processes of the cells promoted by intra-reticular enzymes.



Golgi apparatus has membranes similar to the agranular endoplasmic reticulum. It functions in close relation to the endoplasmic reticulum and is composed of four or more stacked layers of thin, flat, enclosed vesicles lying in proximity of the nucleus. This apparatus is present in abundance in secretory cells on the side from where the secretory substances are extruded.



Waste Disposal: Lysosomes and Proteasomes


The cells depend primarily on lysosomes to degrade internalized material and accumulated internal waste. There are, however, multiple other routes to degrade intracellular macromolecules.


Lysosomes: They are vesicular organelles formed by the disintegration of the Golgi body and subsequent dispersion in the cytoplasm. The lysosomes permit the cell to digest:




  1. 1. Damaged cellular structures.
  2. 2. Food particles that are ingested by the cell.
  3. 3. Unwanted matter such as bacteria.

Hydrolytic enzymes are highly concentrated in lysosomes which function to split the organic substances into smaller substances like amino acids and glucose which facilitates higher rate of diffusion.


Peroxisomes: Similar in structure to lysosomes are peroxisomes. Although lysosomes and peroxisomes differ in two critical ways, peroxiomes are formed by self-replication or budding from the smooth endoplasmic reticulum instead of the Golgi body. Second, they contain oxidases rather than hydrolases. Many oxidases can form hydrogen peroxide (H2O2) by combining oxygen with hydrogen ions derived from different intracellular chemicals.



Cellular Metabolism and Mitochondrial Function


The mitochondria are known as the ‘powerhouses’ of the cell. Without them, cells will not be able to extract enough energy from the nutrients. Consequently, all cellular functions would cease. It comprises two lipid bilayers: an outer membrane and an inner membrane. The infoldings of the inner membrane form shelves onto which oxidative enzymes are attached. These shelves are called cristae and serve as a platform for chemical reactions. The matrix forms the inner cavity of the mitochondrion, which contains large quantities of dissolved enzymes that are essential for deriving energy from nutrients. These enzymes cause oxidation of the nutrients by operating in association with the oxidative enzymes on the cristae. Carbon dioxide and water are the by-products. There is the simultaneous release of energy. The released energy synthesizes a ‘high-energy’ substance called adenosine triphosphate (ATP). ATP is then carried out of the mitochondrion, where it diffuses throughout the cell to release its energy for performing cellular functions.


The nucleus is the cell’s control centre. It sends messages to the cell to grow and mature participate in cell replication, or die. It contains large quantities of D.N.A., which together form the genes are responsible for the various cell protein characteristics. These cell proteins include both the structural proteins and the intracellular enzymes that in turn control the cytoplasmic and nuclear activities. Another important function of the genes is to control and promote the reproduction of the cell by initial replication to make two identical sets of genes. Every cell duplicates by mitosis to produce two daughter cells. Each daughter cell receives one set of D.N.A. genes. The nuclear membrane comprises two separate layers namely inner and outer. The outer nuclear membrane is contiguous with the membranes of the endoplasmic reticulum while the space between the two nuclear membranes continues into the space inside the endoplasmic reticulum.



Cellular Activation


Cell Signalling: The signals that most cells respond to can be classified into the following groups: Danger and pathogens, cell–cell contacts mediated through adhesion molecules and/or gap junctions, cell-ECM contacts mediated through integrins, secreted molecules.


Signalling pathways can also be classified based on the spatial relationships between the sending and receiving cells: Paracrine signalling, autocrine signalling, synaptic signalling, endocrine signalling. All extracellular stimuli (i.e., paracrine, autocrine, synaptic, or endocrine) convey their signal to the cell via specific receptor proteins. Receptors may be present on the cell surface, generally transmembrane proteins with extracellular domains that bind activating ligands. In addition, receptors may be present within the cell, termed intracellular receptors, including transcription factors activated by lipid-soluble ligands that can easily transit plasma membranes. Vitamin D and steroid hormones that activate nuclear hormone receptors are good examples.


Signal Transduction Pathways: The interaction of a cell-surface receptor and its ligand activates signalling through ligand-induced clustering of the receptor (receptor cross-linking) or by inducing a physical change in receptor structure. These mechanisms result in a conformational change in the receptor’s cytosolic tail, resulting in additional intracellular biochemical events.


Modular Signalling Proteins, Hubs and Nodes: Any initial signal that relies on enzymatic activity impacts multiple processes, each is contributing to the outcome, with adapter proteins playing a pivotal role in organizing intracellular signalling pathways. For example, specific phosphorylation of any given protein allows it to associate with a host of other molecules, thereby causing various processes such as enzyme activation (or inactivation), nuclear (or cytoplasmic) localization of transcription factors, activation or inactivation of transcription factor, polymerization (or depolymerization) of actin, degradation or stabilisation of proteins, activation of feedback inhibitory or stimulatory loops.


Transcription Factors: Most signal transduction pathways ultimately induce long-lasting effects on cellular function by modulating gene transcription; this occurs through the activation and/or nuclear localization of transcription factors. Transcription factors often contain:




  • Modular domains that bind to D.N.A
  • Small molecules such as steroid hormones
  • Intracellular regulatory proteins.

Interactions mediated by these domains can be controlled by posttranslational modifications such as phosphorylation. These changes can result in translocation from the cytoplasm into the nucleus, modify transcription factor protein half-life, expose specific D.N.A. binding motifs, or promote binding to components of the R.N.A. polymerase complex to augment transcription factor activity.



Growth Factors and Receptors


Growth factors stimulate signalling pathways and genes that augment cell survival, growth, and division. Binding of the growth factors to specific receptors ultimately influences the expression of genes leading to various processes such as:




  • Promoting the entry into the cell cycle.
  • Relieving the blocks on cell cycle progression leading to promoting of replication.
  • Preventing apoptosis.
  • Enhancing synthesis of nucleic acids, proteins, lipids, carbohydrates which are required for progression of cell division such as epidermal growth factor (EGF), transforming Growth Factor-α (TGF-α), hepatocyte Growth Factor (H.G.F.), vascular Endothelial Growth Factor (VEGF), platelet-derived Growth Factor (PDGF), fibroblast Growth Factor (FGF) and transforming Growth Factor β (TGF-β) (Table of the growth factors from Robbins 10th edition, pg 20).

Extracellular matrix


The E.C.M. comprises a protein network that constitutes a large proportion of any tissue. Therefore, cell interactions with the E.C.M. are critical for developing, healing and maintaining standard tissue architecture.


Apart from the primary function of ‘space-filling’ around the cells, the E.C.M. also serves as a:




  • Mechanical support for cell anchorage, cell migration and maintenance of cell polarity.
  • Regulator of cell proliferation by binding and displaying growth factors and signalling via cellular integrin family receptors, providing a depot for latent growth factors activated within foci of injury or inflammation.
  • Scaffolding for tissue renewal considering maintenance of normal tissue structure requires a basement membrane or stromal scaffolds. Organized tissue regeneration requires the integrity of the basement membrane and the stroma of parenchymal cells.
  • Foundation for establishing tissue microenvironments, serving as a boundary between the epithelium and underlying connective tissue by not just providing structural support; for instance, it forms part of the filtration apparatus in the renal system.

The E.C.M. occurs in two primary forms:


Interstitial matrix and basement membrane

E.C.M. components can be further categorized as (Fig. 2.15.1.3):




  • Fibrous structural proteins (like collagen and elastin) provide tensile strength and recoil.
  • Water-hydrated gels (like proteoglycans and hyaluronan) provide compressive resistance and lubrication.
  • Adhesive glycoproteins that connect E.C.M. elements and to cells.

Image
Fig. 2.15.1.3 Extracellular matrix (ECM) components. (A) Fibrillar collagen and elastic tissue structures. Due to rodlike fibril stacking and extensive lateral cross-linking, collagen fibers have marked tensile strength but do not have much elasticity. Elastin is also cross-linked but differs in having large hydrophobic segments that form a dense globular configuration at rest. As stretch is exerted, the hydrophobic domains are pulled open, but the cross-links keep the tissue intact and release of the stretch tension allows the hydrophobic domains of the proteins to refold. (B) Proteoglycan structure. The highly negatively charged sulfated sugars on the proteoglycan “bristles” attract sodium and water to generate a viscous but compressible matrix. (C) Regulation of basic fibroblast growth factor (bFGF, also known as FGF-2) activity by ECM and cellular proteoglycans. Heparan sulfate binds bFGF secreted in the ECM. Syndecan is a cell surface proteoglycan with a transmembrane core protein and extracellular glycosaminoglycan side chains that can bind bFGF and a cytoplasmic tail that interacts with the intracellular actin cytoskeleton. Syndecan side chains bind bFGF released from damaged ECM, thus facilitating bFGF interaction with cell-surface receptors. FGF, Fibroblast growth factor. Source: (Reproduced from Vinay Kumar, Abul Abbas, Jon Aster. Robbins & Cotran Pathologic Basis of Disease, 10e, The Cell as a Unit of Health and Disease, Figure 1.15, Philadelphia (PA), Elsevier Inc., 2021.)

Maintaining cell population


Proliferation and the cell cycle


Cell proliferation plays a primary role in organism development for maintaining steady-state tissue homeostasis and replacing dead or damaged cells.


It comprises of the following two consecutive processes:




  • D.N.A. replication,
  • Segregation of replicated chromosomes into two separate cells.

Initially, the cell division process was studied into two subheadings:




  • mitosis (M), which is the process of nuclear division which occurs in following stages: prophase, metaphase, anaphase and telophase.
  • interphase, the interlude between 2 M phases. Under the microscope, interphase cells simply appear to grow in size. But different techniques have revealed that the interphase comprises three phases – G1, S and G2. Replication of D.N.A. occurs essentially in the S phase of the interphase. S phase is preceded by the G1 phase during which the cell prepares itself for D.N.A. synthesis. G2 phase follows the S phase during which the cell prepares for mitosis.

Cells in G1 can also enter a resting state called the G0 phase which accounts for the non-growing, non-proliferating cells in the human. Each stage requires completing the previous step and subsequent activation of necessary factors. Non-fidelity D.N.A. replication or cofactor deficiency leads to arrest at different transition points.


Cyclin and Cyclin-dependent kinases (CDKs): The cell cycle is regulated by the activators and inhibitors. Cell cycle is supervised mainly by:




  • Cyclins are proteins named for the cyclic nature of their production and degradation.
  • Cyclin-associated enzymes called cyclin-dependent kinases (CDKs) which acquire the ability to phosphorylate protein substrates by forming complexes with the relevant cyclins.

Cyclins are found in every cell throughout all eukaryotes. The cyclin family is quite largewith 30 identified members to date (N.H. Lents, J.J. Baldassare, in Encyclopedia of Cell Biology, 2016). Different cyclins function in different cell cycle phases along with their associated CDK, and different subtypes show specific tissue-distribution patterns in multicellular organisms. The cyclins D, E, A and B appear sequentially during the cell cycle and bind to at least one or more CDKs. The cell cycle thus resembles a relay during which each leg is regulated by a definite set of cyclins with a different set of cyclins taking charge one after the other. In addition, surveillance mechanisms primed to sense D.N.A. or chromosomal damage are embedded within the cell cycle. These quality control checkpoints ensure that cells with genetic aberrations cannot complete replication. Thus, the G1-S checkpoint monitors D.N.A. integrity before irreversibly committing cellular resources to D.N.A. replication. Later on, during the cell cycle, the G2-M restriction point ensures that there has been precise genetic replication before the cell divides. When cells detect D.N.A. irregularities, checkpoint activation delays cell cycle progression and triggers D.N.A. repair mechanisms. If the genetic abnormality is severe enough not to be repaired, cells either undergo apoptosis or enter a non-replicative state called senescence primarily through p53-dependent mechanisms. Transiently increased synthesis of a particular cyclin thus results in increased kinase activity of the CDK binding partner. As the CDK completes its round of phosphorylation, the associated cyclin is degraded, and CDK activity stops. Consequently, as cyclin levels rise and fall, the activity of associated CDKs will likewise increase and decrease (Fig. 2.15.1.4).


Image
Fig. 2.15.1.4 Therapeutic targeting of the hallmarks of cancer. Drugs that interfere with each of the hallmark capabilities and hallmark-enabling processes have been developed and are in preclinical and/or clinical testing, and in some cases, approved for use in treating certain forms of human cancer. A focus on antagonizing specific hallmark capabilities is likely to yield insights into developing novel, highly effective therapeutic strategies. HGF, hepatocyte growth factor; PARP, poly (ADP-ribose) polymerase; VEGF, vascular endothelial growth factor. Source: (Modified from: Singh, Yatendra & Dubey, Anubhav & Prakash, Om & Tiwari, Deepanshi. (2021). Drug repurposing in Oncology: Opportunities and challenges. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 09. 68-87.)

CDK inhibitors (CDKIs) are responsible for enforcing the cell cycle checkpoints by modulation of CDK-cyclin complex activity. There are several different CDKIs, namely,




  • One family of CDKIs composed of three proteins called p21 (CDKN1A), p27 (CDKN1B) and p57 (CDKN1C) broadly inhibit multiple CDKs.
  • Another family of CDKIs has selective effects on cyclin CDK4 and cyclin CDK6. The proteins involved are p15 (CDKN2B), p16 (CDKN2A), p18 (CDKN2C) and p19(CDKN2D).
  • Defective CDKI checkpoint proteins allow damaged D.N.A. to divide, leading to mutated daughter cells in danger of malignant transformation.

In cancer, unrestrained cell proliferation is achieved by fundamental alterations in the genetic control of cell division by mutations occurring either in protooncogenes and/or in tumour suppressor genes. In normal cells, the products of protooncogenes act at diverse levels along the pathways that stimulate cell proliferation. Mutated forms of protooncogenes or oncogenes can promote tumour growth. Inactivation of tumour suppressor genes like p53 and pRb leads to the dysfunction of proteins that generally inhibit cell cycle progression. Cell cycle deregulation with cancer occurs through mutation of proteins essential at various levels of the cell cycle. Cancers are associated with mutations in genes encoding CDK, cyclins, CDK-activating enzymes, C.K.I., CDK substrates and checkpoint proteins. Many chemotherapeutic therapies use drugs presently either in clinical use or under development, target G1 CDKs, other cell cycle kinases (A.T.M. and A.T.R., CHK1 and CHK2, WEE1, Auroras, polos, MPS1), D.N.A. damage sensors (PARP), kinesins and epigenetic regulators (SETD2 methylase). They are being combined with one another or with conventional cytotoxic agents to advance cancer therapy (Fig. 2.15.1.5).


Image
Fig. 2.15.1.5 Review of regulation and deregulation in cell proliferation. Source: (Adapted from: Vermeulen K, Van Bockstaele DR, Berneman ZN. The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer. Cell proliferation. 2003 Jun;36(3):131-49.)

Stem cells


Stem cells possess self-renewing properties and further differentiate into specific cells and tissues. Although some researchers separate stem cells into multiple subsets, there are fundamentally only two varieties: embryonic stem cells and tissue stem cells.




  • Embryonic stem (E.S.) cells are the most undifferentiated. They constitute within the inner cell mass of the blastocyst, have virtually limitless cell renewal capacity and can differentiate into any cell of the body thus termed as totipotent. While E.S. cells can be maintained for extended periods without differentiating, they can be induced to form specialized cells of all three germ cell layers under appropriate culture conditions.
  • Tissue stem cells/adult stem cells are found in close knit association with the differentiated cells of a given tissue. There are specialized tissue microenvironments called stem cell niches where they are protected. Many organs, particularly the bone marrow, where hematopoietic stem cells congregate in perivascular niches, and the intestines, where epithelial stem cells are confined to the crypts contribute to these stem cells. The bulge region of hair follicles, the cornea’s limbus, and the brain’s subventricular zone constitute other stem cell niches. Soluble factors and other cells within the niches are responsible for maintaining the balance between stem cell quiescence, expansion and differentiation.

Regenerative medicine


The burgeoning field of regenerative medicine is based on identifying, isolating, expanding and transplant stem cells. It should, however, be remembered that this summary is intentionally brief, and one can readily find detailed information about topics reviewed here in textbooks and online literature devoted to cell and molecular biology.


Introduction to cancer


Cancer is one of the leading causes of death in India. While globally, non-communicable diseases (NCDs) accounted for 71% of total deaths, NCDs were estimated to account for 63% of all deaths, with cancer contributing to around 9% of total deaths in India. The projected number of patients with cancer in India was 1,392,179 for 2020. The five leading cancer sites are breast, lung, mouth, cervix uteri, and tongue (Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2018 Nov.; 68(6):394–424.) make a reference out of this in the end.


Even more excruciating than the mortality associated with cancers is the emotional and physical trauma inflicted. Some cancers, like Hodgkin lymphoma, are curable, whereas others, like pancreatic adenocarcinoma, are virtually always fatal. The only hope for controlling cancer lies in learning more about its causes and pathogenesis. Fortunately, great strides are constantly being made in understanding its molecular basis. Some excellent study results have emerged: cancer mortality for both men and women in the United States has declined during the last decade of the 20th century. It has continued its downhill course in the 21st century. The use of routine radiographic imaging provides a promising platform for linking specific imaging traits with respective gene expression patterns, further highlighting the underlying cellular pathophysiology. The imaging features assist molecular surrogates that contribute to the diagnosis, prognosis, and gene-expression-associated treatment response associated with diverse forms of human cancer. Gene expression profiling relies on the surgical procurement of tissue, involving several risks and potential complications and making it an impractical option for every cancer patient. Radiographic imaging studies in contrast are part of routine clinical care. It is mainly used to gather important anatomical and morphological information and is not perceived to imply much consequential molecular detail. Methods that assist in analyzing lesion characteristics such as permeability and blood flow, like dynamic contrast-enhanced (D.C.E.) magnetic resonance imaging (M.R.I.) or dynamic computerized tomography, are equipped to distinguish benign from malignant lesions. These can define the topographical boundaries of the cancer lesion, thereby improving staging accuracy.


Nomenclature of cancer


Neoplasia means ‘new growth’ in Greek, and thereby, a collection of cells and stroma composing new growths is termed a neoplasm. The tumour was described initially as swelling caused by inflammation but is now equated with neoplasm. Oncology (Greek oncos = tumour) is defined as the study of tumours or neoplasms. In the modern era, a neoplasm is defined as a genetic disorder of cell growth triggered by acquired or less commonly inherited mutations affecting a single cell and its clonal progeny.


All tumours are composed of following main components:




  1. (1) Neoplastic cells that constitute the tumour parenchyma.
  2. (2) Reactive stroma which is made up of connective tissue, blood vessels and cells of the adaptive and innate immune system.

Classification of cancer: Benign vs malignant vs mixed





  • Benign tumours are the ones that remain localized to their site of origin and are generally amenable to surgical removal. Predictably, the patient typically has a good prognosis. The naming of benign tumours of mesenchymal cells is comparatively simple; most commonly, the suffix ‘-oma’ is attached to the name of the cell type from which the tumour arises, like fibroma, chondroma. The nomenclature of benign epithelial tumours is more complex; they are classified either based on their cell of origin, microscopic appearance or macroscopic architecture.
  • Malignant tumours have the ability to invade and destroy the adjacent structures and spread/metastasize to distant sites. Malignant tumours are labelled as cancers, derived from the Latin word for crab because they tend to cling onto any part obstinately. Malignant tumours originating from solid mesenchymal tissues are usually called sarcomas (Greek sar = fleshy; e.g., fibrosarcoma and chondrosarcoma). Those arising from blood-forming cells are termed leukaemias (literally, white blood) or lymphomas (tumours of lymphocytes or their precursors). Malignant neoplasms of epithelial origin are called carcinomas.
  • Mixed Tumours, unlike most neoplasms where all parenchymal cells closely resemble each other, some tumours demonstrate more than one line of differentiation, thereby creating unique subpopulations of cells. A classic example of a mixed tumour is the exocrine gland, which contains epithelial components scattered within a myxoid stroma further characterized by islands of cartilage or bone. Many neoplasms, including mixed tumours, are composed of cells derived from a single germ layer (i.e., either mesoderm, endoderm or ectoderm). An exception is a teratoma, which contains recognizable mature or immature cells or tissues belonging to more than one germ cell layer (and sometimes all three) (Table 2.15.1.2).


TABLE 2.15.1.2


Nomenclature of Tumors











































































































Tissue of Origin Benign Malignant
COMPOSED OF ONE PARENCHYMAL CELL TYPE
TUMORS OF MESENCHYMAL ORIGIN

Connective tissue and derivatives


Fibroma


Lipoma


Chondroma


Osteoma


Fibrosarcoma


Liposarcoma


Chondrosarcoma


Osteogenic sarcoma

VESSELS AND SURFACE COVERINGS

Blood vessels


Hemangioma


Angiosarcoma


Lymph vessels


Lymphangioma


Lymphangiosarcoma


Mesothelium


Benign fibrous tumor


Mesothelioma


Brain coverings


Meningioma


Invasive meningioma


Blood Cells and Related Cells


Hematopoietic cells



Leukemias


Lymphoid tissue



Lymphomas


Muscle


Smooth


Leiomyoma


Leiomyosarcoma


Striated


Rhabdomyoma


Rhabdomyosarcoma

TUMORS OF EPITHELIAL ORIGIN

Stratified squamous


Squamous cell papilloma


Squamous cell carcinoma


Basal cells of skin or adnexa



Basal cell carcinoma


Epithelial lining of glands or ducts


Adenoma


Papilloma


Cystadenoma


Adenocarcinoma


Papillary carcinomas


Cystadenocarcinoma


Respiratory passages


Bronchial adenoma


Bronchogenic carcinoma


Renal epithelium


Renal tubular adenoma


Renal cell carcinoma


Liver cells


Hepatic adenoma


Hepatocellular carcinoma


Urinary tract epithelium (transitional)


Transitional cell papilloma


Transitional cell carcinoma


Placental epithelium


Hydatidiform mole


Choriocarcinoma


Testicular epithelium (germ cells)



Seminoma


Embryonal carcinoma


Tumors of Melanocytes


Nevus


Malignant melanoma

MORE THAN ONE NEOPLASTIC CELL TYPE—MIXED TUMORS, USUALLY DERIVED FROM ONE GERM CELL LAYER

Salivary glands


Pleomorphic adenoma (mixed tumor of salivary origin)


Malignant mixed tumor of salivary gland origin


Renal anlage



Wilms tumor

MORE THAN ONE NEOPLASTIC CELL TYPE DERIVED FROM MORE THAN ONE GERM CELL LAYER—TERATOGENOUS

Totipotential cells in gonads or in embryonic rests


Mature teratoma, dermoid cyst


Immature teratoma, teratocarcinoma


Reproduced from Vinay Kumar, Abul Abbas, Jon Aster. Robbins & Cotran Pathologic Basis of Disease: South Asia Edition, Neoplasia, Table 7.1, New Delhi, Elsevier India, 2014.


Cancers can be further characterized in terms of





  • Anaplasia and differentiation: Differentiation refers to how closely the neoplastic cells resemble normal parenchymal cells, both morphologically and functionally. A lack of differentiation is termed anaplasia. Most commonly, benign tumours are well-differentiated whereas most malignant neoplasms exhibit morphologic alterations that betray their potential for aggressive behaviour. In addition to anaplasia, cancer cells often show other characteristic morphologic changes such as pleomorphism, abnormal nuclear morphology, mitoses and loss of polarity.


    • Metaplasia, Dysplasia, and Carcinoma in situ: Metaplasia is defined as the replacement of one type of cell line with another kind. It is mainly associated with tissue damage, repair and regeneration. Dysplasia means a ‘disordered growth’, encountered principally in epithelial cells and is recognized based on several morphologic changes. When dysplasia is severe and involves the entire epithelium thickness, but the lesion does not invade the basement membrane, it’s labelled carcinoma in situ.

  • Local invasion: The growth of cancers is associated with progressive invasion, destruction of surrounding tissue and eventually systemic spread. Nearly all benign tumours grow as cohesive, expansile masses that remain localized to their site of origin and cannot invade or metastasize to distant sites. After metastases, invasiveness is the next most reliable discriminator of malignant and benign tumours.
  • Metastasis: Metastasis is the spread of a tumour to physically distant sites from the primary site of the tumour, which unequivocally marks a tumour as malignant.
  • Pathways of Spread: Dissemination of cancers occurs through three pathways, namely, (1) direct seeding of body cavities or surfaces, (2) lymphatic spread, and/or (3) hematogenous spread.

Molecular basis of cancer: Role of genetic and epigenetic alterations


Non-lethal genetic damage is the major cause of carcinogenesis. A tumour is formed due to clonal expansion of one precursor cell that has incurred genetic damage (i.e., tumours are clonal). The principal targets of cancer-causing mutations are four classes of genes that regulate programmed necrosis (apoptosis). These genes are liable for D.N.A. repair, growth-promoting protooncogenes and growth-inhibiting tumour suppressor genes. Carcinogenesis results from the cumulation of mutations over time. Once established, tumours evolve genetically during their growth and progression under the pressure of Darwinian selection (survival of the fittest). As predicted, these studies identified two kinds of mutations were identified: (1) mutations that are present altogether tumour sites tested, which were presumably present in the cell of origin at the instance of transformation and (2) mutations that are unique to a subset of tumour sites, which were likely acquired after transformation during the outgrowth and spread of the tumour. The natural transformation of cancer and changes in tumour behaviour following therapy abide by the theory of the selection of the fittest cells. In addition to D.N.A. mutations, epigenetic aberrations also contribute to the malignant properties of cancer cells. Epigenetic modifications include D.N.A. methylation, which silence gene expression, and modifications of histones, the proteins that package D.N.A. into chromatin, which may enhance or dampen genetic expressivity.


Cellular and molecular hallmarks of cancer (Fig. 2.15.1.4)


Self-Sufficiency in Growth Signals


Oncogenes are mutated genes that cause excessive cell growth, even without growth factors and other growth-promoting external cues. A significant discovery in cancer was that oncogenes are mutated or overexpressed versions. Physiologic growth factor-induced signalling is studied mainly under following headlines:




  • Growth factor binding to its specific receptor.
  • Transient and limited activation of the growth factor receptor with subsequent activation of several cytoplasmic signal-transducing proteins.
  • Transmission of the transduced signal to the nucleus via additional cytoplasmic effector proteins and second messengers or by a cascade of signal transduction molecules.
  • Induction and activation of transcription factors and epigenetic alterations that initiate and sustain D.N.A. transcription.

Aberrations in multiple signalling pathways have been observed to play a significant role in the formation of neoplasms. Components of these pathways serve as oncoproteins when mutated. Whereas normal cells regulate cell growth and division to maintain cell homeostasis, cancer cells can frequently proliferate. The assembly of protein ligands accomplishes proliferation. These send signals to normal cells to supply growth factors, increasing receptor proteins on the neoplastic cell surface to make them sensitive to growth factor ligands. Epidermal growth factor receptor (EGFR) is a trans-membrane glycoprotein with two main components, an extracellular epidermal growth factor binding domain and an intracellular tyrosine kinase domain that controls signaling pathways regulating cellular proliferation. Upon binding of a specific ligand, there is conformational change in the normally functioning EGFR and phosphorylation of the intracellular domain occurs, causing downstream signal transduction by various pathways such as the Raf1-extracellular signal-regulated kinase, PI3K/Akt, and signal transducer and activator of transcription (STAT) factors. Depending on the pathway, there is inhibition of apoptosis withthe common end result being cell proliferation or cell maintenance. Overexpression of EGFR or mutations in intracellular EGFR has been observed in 43%–89% cases of non-small cell lung cancer (Bethune G et al, Journal of thoracic disease, 2010).


c-Kit, a receptor tyrosine kinase, is involved in intracellular signalling. The mutated form of c-Kit plays a significant role in the occurrence of some cancers, leading to the concept that inhibiting c-Kit kinase activity can be a target for cancer therapy (Babaei MA, Drug design, development and therapy, 2016). After ligand binding, K.I.T. causes phosphorylation and subsequent activation of downstream members of the Ras-Raf-MAPK, Jak/Stat, and PI3k/A.K.T. signalling pathways, thus, controlling cell proliferation, apoptosis, chemotaxis, and metabolism. Gain of function mutations in K.I.T. is present in 55% of gastrointestinal stromal tumours (GISTs). EGFR may be a tyrosine kinase receptor comprising four receptors: ErbB1 (EGFR/HER1), ErbB2, ErbB3, and ErbB4 (HER4). E.G.F.R. is overexpressed in several human tumours, including lung, breast, colorectal, and head and neck cancers. In a phenomenon termed oncogene addiction, N.S.C.L.C. tumours require mutant, E.G.F.R.S for proliferation and survival. Erlotinib, gefitinib, and afatinib are, for E.G.F.R. Tyrosine Kinase Inhibitors approved by the U.S. Food and Drug Administration (F.D.A.) as first-line therapy for N.S.C.L.C. with, E.G.F.R. mutations. The initial clinically significant decrease in tumour burden reflects the attribute of oncogene addiction to EGF and forms the basis of the dramatic response to the EGFR TKI therapy. Although after a median of 12 months, patients who initially demonstrated a good response to, for E.G.F.R. inhibitors usually experience slow tumour progression due to resistance. This resistance is caused due to a second-site mutation T790M at exon 20 in approximately one-half of total cases. Skin toxicity, colitis and rare but potentially fatal is pneumonitis are the main toxicities commonly associated with, E.G.F.R. inhibitors. On C.T. scan this pneumonitis is seen as multifocal ground-glass opacities with or without interlobular septal thickening or diffuse ground-glass densities with consolidation and traction bronchiectasis and are most often misdiagnosed as tumour spread in patients with N.S.C.L.C.


The anti-EGFR–directed therapies have been effectively used in other solid malignancies, such as colorectal, head and neck, and breast cancer for instance anti-EGFR antibodies such as cetuximab and panitumumab are presently approved to treat colorectal cancer and head and neck squamous cell cancer. Infusion reaction and rash are a few of the toxicities seen with the anti-EGFR monoclonal antibodies. The acneiform rash seen with cetuximab correlates with a greater response rate in carcinoma treatment. Trastuzumab is a humanized antibody against the extracellular domain of EGFR/HER2 which is commonly seen in breast, gastric and gastroesophageal junction cancers and it causes down-regulation of the intracellular PI3K and M.A.P.K. signalling pathways thereby activating the immune response. Other HER2-targeting agents utilized in carcinoma are pertuzumab, trastuzumab-emtansine, and lapatinib. Pertuzumab inhibits dimerization at HER2/HER3. Pertuzumab shows improved blockade of the HER signalling pathway when combined in treatment with trastuzumab. Cardiotoxicity associated with trastuzumab manifests on imaging as cardiomegaly, pleural effusions and interlobular septal thickening. Imatinib is a small-molecule T.K.I. inhibiting KIT. Toxicities of imatinib manifest on imaging as fluid retention which is seen as ascites, pleural effusions, pulmonary oedema and anasarca.



Oncoproteins and Cell Growth


Oncogenes have multiple roles, but virtually all encode constitutively active oncoproteins that participate in signalling pathways that regulate the proliferation of cells. Thus protooncogenes, the normal regulated versions of oncogenes, may encode growth factors, growth factor receptors, signal transducers, transcription factors or cell cycle components.




  • Growth Factors: Most growth factors are made by 1 cell type and act on a neighbouring cell of a differing kind expressing the appropriate growth factor receptor (paracrine action). Many cancer cells synthesize the same growth protein they respond to, thereby creating an autocrine loop. For example, brain tumours such as glioblastomas often express PDGF and PDGF receptor (P.D.G.F.R.).
  • Growth Factor Receptors. Many oncogenes encode protein receptors, of which receptor tyrosine kinases are arguably the most critical in cancer. Recall that receptor tyrosine kinases are transmembrane proteins with an extracellular growth factor-binding domain and a cytoplasmic tyrosine kinase domain. The oncogenic versions of those receptors are related to mutations that cause constitutive, growth factor-independent tyrosine kinase activity. Receptor tyrosine kinases are activated in tumours by multiple mechanisms, including point mutations, gene rearrangements and gene amplification.
  • Downstream Components of the Receptor Tyrosine Kinase Signaling Pathway: Point mutations of R.A.S. family genes constitute the most commonly observed abnormality.
  • Non-receptor tyrosine kinases: Oncogenic mutations are known to also occur in several nonreceptor tyrosine kinases that normally localize to the cytoplasm or the nucleus. In many instances, the mutations form chromosomal translocations or rearrangements, thereby creating fusion genes encoding constitutively active tyrosine kinases. Despite their non-membranous localization, these oncoproteins seem to activate the same signalling pathways as receptor tyrosine kinases. Malignant mesothelioma, pleural effusion (cell block): round shaped cells, often binucleated, cytoplasmic vacuolatuion (H&E, 40x). In chronic myeloid leukaemia and a subset of acute lymphoblastic leukaemia, the A.B.L. gene is translocated from its normal location on chromosome nine to chromosome 22.
  • Transcription factor: Almost all the signal transduction pathways converge on the nucleus. This leads to the ultimate result of deregulated mitogenic signalling pathways and continuous stimulation of nuclear transcription factors that drive growth-promoting genes. Growth autonomy may also occur due to mutations of transcription factors thereby up-regulating the expression of pro-growth genes and cyclins. This class includes the M.Y.C., M.Y.B., Jun., F.O.S. and R.E.L. protooncogenes.
  • Cyclins and Cyclin-Dependent Kinases: CDKs which are activated by binding to cyclins are responsible for the cell cycle regulation. They are termed after the cyclic nature of their production and degradation. Phosphorylation of the crucial target proteins is carried out by the CDK-cyclin complexes that drive cells forward through the cell cycle. While cyclins arouse the CDKs, CDK inhibitors, of which there are many, silence the CDKs and exert negative control over the cell cycle. The major cancer-associated mutations that affect the G1/S checkpoint can be broadly grouped into two classes:


    • Gain-of-function mutations in D cyclin genes and CDK4 promote unregulated G1/S progression and thus function as oncogenes.
    • Loss-of-function mutations in genes that inhibit G1/S progression.

Insensitivity to Growth-Inhibiting Signals


Tumour suppressor proteins function to control a series of checkpoints responsible for preventing uncontrolled growth. Many tumour suppressors, such as R.B. and p53, constitute a regulatory network that recognizes genotoxic stress from any source and responds by halting proliferation. The protein products of tumour suppressor genes thereby function as transcription factors, cell cycle inhibitors, signal transduction molecules, cell surface receptors and cellular response regulators to D.N.A. damage.




  • RB, Governor of the cell cycle: R.B. hypophosphorylation creates antiproliferative effects by binding and inhibiting E2F transcription factors. These transcription factors regulate genes and thereby help the cells to pass successfully through G1/S phase cell cycle checkpoint. Usual growth factor signalling results in R.B. hyperphosphorylation and inactivation, thus encouraging cell cycle progression. The antiproliferative effect of R.B. is annulled in cancers through different mechanisms, including loss-of-function R.B. mutations, amplification of the CDK4 and cyclin D genes, loss-of-function mutations affecting CDK inhibitors (e.g., p16/INK4a), viral oncoproteins that bind and inhibit R.B. (E7 protein of HPV).
  • P53, guardian of the genome: The p53 protein functions as the central monitor of stress in the cell and is activated by stimuli such as anoxia, inappropriate signalling by mutated oncoproteins, or D.N.A. damage. p53 regulates the expression and activity of proteins involved in cellular physiology, such as cell cycle arrest, D.N.A. repair, cellular senescence, and apoptosis. D.N.A. damage is perceived by complexes containing ATM/ATR family kinases. P53 is liberated from inhibitors such as MDM2 upon phosphorylation by these kinases. Cell-cycle arrest at the G1/S checkpoint is then manifested indirectly by activated p53 that upregulates the expression of proteins such as the CDK inhibitor p21. This pause allows cells to repair D.N.A. damage. If D.N.A. damage cannot be repaired, p53 induces additional events that lead to cellular senescence or apoptosis. A large number of human cancers demonstrate biallelic loss-of-function mutations in TP53. Patients with rare Li-Fraumeni syndrome inherit one defective copy of TP53 and have a relatively high incidence of various cancers. Like R.B., p53 is inactivated by viral oncoproteins, such as the E6 protein of HPV.

Mechanism of Action of Major Tumour Suppressor Genes




  • A.P.C.: It functions to encode a factor that downregulates the WNT pathway in colonic epithelium by forming a complex responsible for the degradation of β-catenin. Familial adenomatous polyposis, an autosomal dominant disorder associated with the development of thousands of colonic polyps and early-onset colon carcinoma, is formed due to germline loss-of-function mutations. Acquired somatic A.P.C. mutations are demonstrated in about 70% of sporadic colon carcinomas.
  • E-cadherin: It is a cell adhesion molecule and plays a crucial role in contact-mediated growth inhibition of epithelial cells. It also regulates the WNT pathway by binding and sequestering of β-catenin, a signalling protein that functions in the pathway. Autosomal dominant familial gastric carcinoma is caused due to germline loss-of-function mutations in the E-cadherin gene (CDH1). Loss of CDH1 expression is observed in many sporadic carcinomas; these are associated with loss of contact inhibition, loss of cohesiveness, increased invasiveness, and increased WNT signalling.
  • CDKN2A: A complex locus encodes two tumour suppressive proteins, p16/INK4a (CDK inhibitor that augments R.B. function) and A.R.F. (which stabilizes p53). Autosomal dominant familial melanoma is caused due to germline loss-of-function mutations of the same. Biallelic loss of function is observed in various cancers including leukaemias, melanomas and carcinomas.
  • TGF-β pathway: It is a potent inhibitor of cellular proliferation in normal tissues. It hold crucial clinical importance as its loss-of-function mutations involving TGF-β have frequent association with various carcinomas such as colon, stomach, endometrium, while those involving downstream signal transducers lead to S.M.A.D.s and pancreatic carcinomas. This pathway is hypothesized to also have a pro-oncogenic role by enhancing the immune evasiveness of tumours.
  • P.T.E.N. encodes a lipid phosphatase that plays a crucial role as a negative regulator of PI3K/AKT signalling. Germline loss-of-function mutations cause Cowden syndrome, an autosomal dominant disorder relating to a high risk of breast and endometrial cancers. Biallelic loss of function is common in diverse cancers.
  • V.H.L.: It encodes a component of a ubiquitin ligase complex responsible for the degradation of hypoxia-induced factors, which are transcription factors that alter gene expression in response to hypoxia. Germline loss-of-function mutations result in von Hippel–Lindau syndrome, an autosomal dominant disorder associated with the occurrence of renal cell carcinoma and pheochromocytoma. Sporadic renal cell carcinoma usually demonstrates acquired biallelic loss-of-function mutations.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Mar 25, 2024 | Posted by in CARDIOVASCULAR IMAGING | Comments Off on Oncoimaging: Basic science to clinical application

Full access? Get Clinical Tree

Get Clinical Tree app for offline access