CBIO 4: Tissue Invasion and Metastasis Flashcards

You may prefer our related Brainscape-certified flashcards:
1
Q

What are the stages of the metastatic process?

A
  1. Invasion of adjacent tissue by cells from the primary tumour
  2. Vascularisation of the primary tumour and intravasation of cancer cells (entering the blood vessel)
  3. Transport of metastatic cells through the circulatory system.
  4. Cell arrest at a secondary site and extravasation of cancer cells (escaping the blood vessel)
  5. Growth of tumour in a secondary organ/site
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What does it mean clinically when a metastatic tumour is diagnosed?

A
  • it indicates a terminal disease
  • While our knowledge of tumour biology is increasing, and all-stage survival across all cancer types is increasing, the survival of patients with metastatic disease (stage 4) is not improving greatly.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Read the graph below that compares the one year survival rates for breast, colon, lung and ovarian cancers in 2004 – 2007 and 2012.

Which of the following stage 4 cancer types have improved between 2004-2007 and 2012?

A
  • Improved: Breast cancer
  • Not changed: Lung and ovarian cancer
  • According to Public Health England data, the one-year survival rate for stage 4 breast cancer has increased from around 50% in 2004-2007 to around 65% in 2012.
  • However, for other cancer types the survival rate remains largely unchanged.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What are circulating tumour cells (CTCs)?

A
  • these are cancer cells that escape from primary tumours into blood or lymphatic vessels to disseminate to other organs
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Why has screening for CTCs taken so many years to be developed?

A
  • Screening for CTCs is technically challenging.
  • CTCs are rare compared with other circulating cells and there is no universal surface marker to recognise them by.
  • Apart from CTCs, human blood contains other material that can originate from primary tumours, including cell-free tumour DNA (ctDNA) and RNA (ctRNA), proteins, and vesicles (exosomes).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is a liquid biopsy?

A
  • Liquid biopsy is a clinical test to detect circulating tumour cells or tumour-derived material in the blood and other fluids from patients with cancer.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What other fluids apart from blood can you take a liquid biopsy from?

A
  • urine
  • saliva
  • plerual effusions
  • cerebrospinal fluid (CSF)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What can liquid biopsy do?

A
  • it is a minimally invasive method of monitoring of patients, especially when it comes to monitoring tumour evolution over time
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why is screening a liquid biopsy challenging?

A
  • ctDNA (circulating tumour DNA) is fragmented and highly under-represented compared with tumour DNA
  • The small number of CTCs that can be isolated in a blood sample; approximately 1 cell per 1x109 blood cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

In order to detect CTCs via a liquid biopsy, what needs to be done?

A
  • the sample needs to be enriched
  • in other words, the number of CTCs in the sample needs to be increased.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are the two techniques to increase CTCs?

A
  • negative enrichment:
  • removing other blood cells based on shape, size or other biophysical properties
  • it is focused on other cells other than CTCs
  • positive enrichment:
  • increase CTC numbers by selecting cells expressing specific markers on cell surfaces (surface markers)
  • these surface markers can distinguish epithelial cells from blood cells
  • e.g. epithelial cell adhesion molecules (EpCAM)
  • the limitation is these markers do not distinguish between a malignant and non-malignant epithelial cell
  • so isolated CTCs have to be molecularly characterised
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What technologies are used to analyse liquid biopsies?

A
  • Analysis of liquid biopsies requires highly sensitive assays, which have only recently become available.
  • At this time, the only FDA-approved platform for the isolation and enumeration of CTCs in patients with metastatic breast, colorectal, or prostate cancer is the CellSearch platform.
  • CellSearch uses positive enrichment based on positive expression of EpCAM and negative expression of CD45 (leukocyte-specific molecule)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What needs to happen to cancer cells before the invasion-metastasis cascade?

What triggers this? How?

A
  • cancer cells need to be disseminated
  • the initial trigger for this is genomic instability
  • of which chromosome instability (CIN) is present in most human cancers
  • this arises as a result of errors in chromosome segregation during mitosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the fuel behind the metastatic process?

What are the after-effects of this?

A
  • genomic instability is the fuel behind the metastatic process
  • it can create many cell subtypes, called clones, which may have metastatic properties
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What is monoclonal and polyclonal metastasis?

A
  • monoclonal metastasis:
  • metastatic tumours forming from one cell
  • polyclonal metastasis:
  • metastatic tumours forming from multiple cells
  • which form it forms from is still under debate
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Explain this diagram

A
  • a cancer mass can contain two clones of cancer cells, clone A and clone B.
  • From this clone A and clone B mass, a metastasis can be formed containing both clones (polyclonal), only one clone (monoclonal), or one clone which has undergone further genetic alterations (phenotypically heterogenic monoclonal)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

What two roles does angiogenesis play in tumour progression?

A
  1. Intravasation: Entering blood vessels
  2. Extravasation: Escaping blood vessels
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What are pro-angiogenic molecules?

Why are they needed?

A
  • cancer cells release ‘pro-angiogenic’ molecules which promote the formation of new blood vessels
  • cancer cells drive the process of angiogenesis with this
  • while the primary tumour expands in size and invades surrounding tissues, it requires nutrients and oxygen, which needs to be delivered via the blood vessels
  • these are formed around the tumour mass
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Give an example of a key pro-angiogenic molecule

Describe what happens to the blood vessels formed from its production

A
  • vascular endothelial growth factor (VEGF)
  • These new blood vessels have a “leaky” structure and permits the tumour cells to enter the blood stream, a process which you now know is called intravasation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the epithelial to mesenchymal transition (EMT)?

A
  • A key event for stationary tumour cells to escape the tumour mass so they can migrate and invade
  • During EMT, tumour cells gain a mesenchymal phenotype through a transdifferentiation process and gain new properties so they can invade, resist stress and disseminate
  • Transdifferentiation is the conversion of one cell type into another cell type without going through a pluripotent cell state.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Explain this diagram

A
  • EMT completely alters the characteristics and behaviour of tumour cells and as a consequence, the BM is breached and the cells intravasate into blood or lymphatic vessels.
  • They travel through the blood and lymphatic vessels until they reach their new destination, escape from the vessels (extravasation) and migrate into a new tissue to expand a metastatic colony there
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What do EMT-like changes do to nonepithelial tumours?

A
  • Accumulating evidence suggests that EMT-like changes also lead to a gain in mesenchymal properties and promote malignancy of nonepithelial tumours such as melanoma, sarcoma and leukaemia.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

What is a pre-metastatic niche?

A
  • The new tissue microenvironment after invasion provides a site that promotes colonisation of the tumour cells.
  • It can be altered by bone marrow-derived cells or other circulatory factors prior to tumour cell arrival , forming a pre-metastatic niche
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Briefly recap the basal lamina

A
  • The BM is composed of epithelial, endothelial, and stromal cells to separate the epithelium or endothelium from the stroma and interstitial matrix.
  • The BM is a specialised type of extracellular matrix (ECM) as it is more compact, less porous and has a distinctive composition (consisting of type IV collagen, laminins, fibronectin and linker proteins, including nidogen and entactin)
  • In contrast, the interstitial matrix contains fibrillar collagens, proteoglycans, and various glycoproteins, as you can see in the figure below
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

How do tumour mass and metastatic cancer cells alter the composition of the ECM?

A
  • the alterations arise from enzymes called proteases
  • they degrade proteins of the ECM by hydrolysis of peptide bonds.
  • key proteases:
  • aspartic proteases
  • cysteine proteases
  • serine proteases
  • matrix metalloproteinases (MMPs)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Describe cathepsins proteases

Types

Roles

A
  • Cathepsin D (an aspartic protease)
  • cathepsins B, L and H (cysteine proteases)
  • cathepsin A (a serine protease).

Role:

  • Turnover and degradation of the ECM
  • Activation, processing or degradation of various growth factors, cytokines and chemokines
  • Influence cell-cell adhesion molecules
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

Describe Urokinase proteases

A
  • a serine proteases
  • Key player: urokinase receptor, urokinase-type plasminogen activator receptor (uPAR)

Role:

  • Regulation of urokinase proteolytic activity and ECM components degradation;
  • Regulation of cell adhesion, migration, proliferation and survival by interactions with other transmembrane receptors, like integrins.
  • uPAR binds also to vitronectin (a component of provisional ECM), and through this direct interaction, triggers changes in cell morphology, migration and signalling.
  • One consequence is the induction of EMT (more on this later).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Describe MMP proteases

A
  • MMPs can be divided in six groups:
  • collagenases, gelatinases, stromelysins, matrilysins, membrane-type MMPs, and other non-classified MMPs.

Role:

  • Degradation of collagen and other proteins in the ECM;
  • Regulation of cell behaviour
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Label the diagram

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What is the master cell-cell junction?

How does this relate to metastasis?

A
  • the master cell-cell junction is the adherens junction (cadherins), that regulates the function of other cell-cell junctions
  • In order to break away from the tumour, cells must overcome the mechanisms that cause them to adhere to their neighbours – such as cadherins
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

How do integrins play a role in cancer metastasis?

A
  • integrins interact with specific ECM ligands, sensing the outside-in signals and becoming active.
  • Through this, they can sense the ECM alternations and trigger cells to undergo responsive changes.
  • On the other side of the cell membrane, inside the cell, integrins interact with intracellular ligands and recruit focal adhesion kinase (FAK), which undergoes autophosphorylation, formation of complex with Src and activation of both kinases.
  • The FAK/Src complex can now activate a plethora of signalling molecules.
32
Q

What phenotypic changes are observed during the epithelial to mesenchymal transition (EMT)?

What are the key players of this?

A
  • tumour cells undergoing EMT gain an invasive phenotype by losing their epithelial characteristics and acquiring mesenchymal (stromal) characteristics (see diagram)
  • key players are cadherins and integrins
33
Q

Explain in detail how epithelial cells transition to mesenchymal phenotype via EMT

A
  • Epithelial cells express high levels of epithelial markers (e.g. E-cadherin, EpCAM and α6β4 integrins).
  • However, as cells undergo EMT, the progressive loss of epithelial markers result in the loss of cell-cell adhesion complexes, remodelling of the cell-ECM interactions and production of enzymes to break down the BM and facilitate invasion.
  • Increased expression of mesenchymal markers such as N-cadherin.
  • This results in the “cadherin switch” which also alters cell adhesion.
  • These cells lose their association with epithelial cells and gain enhanced affinity for mesenchymal cells through N-cadherin interactions.
  • Changes in the cytoskeleton such as intermediate filament composition results in increased Vimentin expression enabling cancer cells to become motile.
34
Q

What causes changes in gene expression that contribute to the loss of epithelial markers and gain of mesenchymal markers?

A
  • it is due to master regulators of EMT, transcription factors (EMT-TFs):
  • Snail, Slug, Zeb1 and Twist
  • Their expression is activated early in EMT and therefore has a central role in the cancer progression.
  • EMT can be induced by numerous signalling pathways
  • including TGF-β, BMP, Wnt–β-catenin, NOTCH, and receptor tyrosine kinases.
  • TGF-β signalling is the best known inducer of EMT and is secreted by stromal fibroblasts in the ECM
35
Q

Is the EMT programme a binary switch?

A
  • The EMT programme does not operate as a binary switch between fully epithelial and fully mesenchymal states.
  • It is unclear whether there are discrete phenotypic states along the spectrum or whether EMT is a continuum of states with undefined boundaries
  • As shown in the diagram above, it is also a reversible process and cells can undergo mesenchymal to epithelial transition (MET).
  • Not much is known currently but is thought upon arrival at the new site, the cancer cells can undergo MET to form a new carcinoma.
  • During MET cancer cells re-express their epithelial characteristics and proliferate.
36
Q

What is mesenchymal migration?

What allows this to happen?

A
  • Filamentous (F)-actin plays a role in producing forward cell protrusions.
  • These protrusions allow cells to migrate, in a mode called mesenchymal migration, representing cell migration on a basement membrane.
37
Q

What is amoeboid migtation?

A
  • a round or ellipsoid cell that doesn’t adhere strongly to the ECM can travel via amoeboid migration.
  • In amoeboid migration, actin-rich front protrusions are also present, but their production and retraction is rapid
  • During amoeboid migration, non-actin protrusions known as blebs can be also present. These are produced through hydrostatic pressure and cytoplasmic flow.
38
Q

What is epithelial migration?

A
  • Epithelial migration is migration of a group of cells that are connected through cell-cell adhesions.
  • These groups of cells can move as clusters, sheets, strands or fluid-like masses.
  • This process is also termed bulk migration.

The different modes of cell migration are illustrated in the diagram

39
Q

What adaptation do cancer cells acquire to escape from the primary tumour?

A
  • for cancer cells to escape the primary tumour, they need to alter their expression of adhesion molecules: cadherins, which hold together epithelial cells, and integrins, which allow cells to interact with the extracellular matrix
  • This facilitates epithelial to mesenchymal transition, where cancer cells lose their epithelial characteristics, acquiring mesenchymal characteristics and becoming more motile.
40
Q

What is haematogenous spread?

For which cancers is this common?

A
  • when metastatic cancer cells spread using the blood circulation
  • this is common for sarcomas and some carcinomas
41
Q

What is lymphatic spread?

For which cancers is this common?

A
  • when cancer cells spread using lymphatic channels
  • This is a common route for carcinomas (cancers originating in the inner or outer epithelial walls of the body), but not sarcomas (cancers of the mesodermal origin).
42
Q

What route does breast cancer commonly spread via?

A
  • the lymphatic route
  • Lymphatic spread can itself lead to haematogenous spread, as in the case of breast cancer which can spread through the lymph system, but also to the bone marrow through blood circulation.
43
Q

Can you think of any other ways for cancer cells to spread to other parts of our body?

A
  • There are also other routes of metastasis:
  • Transcoelomic
  • Perineural
  • Leptomeningeal.
44
Q

What is transcoelomic spread?

A
  • In transcoelomic metastasis, cancer cells spread through the surfaces of body cavities.
  • For example, ovarian cancer can spread into the peritoneal cavity, and in cases of lung cancer, the cancer can spread into the pleural cavity.
45
Q

What is perineural spread?

A
  • During perineural spread, cancer cells spread into the layers of nerves.
  • This type of spread can be found in the head and neck, prostate and colorectal cancers, which are highly innervated

https://youtu.be/v1-mJgUQ54M

46
Q

What is leptomeningeal spread?

A
  • In leptomeningeal spread, cancer cells travel by the cerebral spinal fluid
  • This is characteristic for cancers occurring in the brain, including non-Hodgkin’s lymphoma.
  • Cancer cells can travel by the cerebral spinal fluid (CSF).
  • They invade the membrane covering the brain (meninges) to form leptomeningeal metastasis within it, or the spinal cord to invade the dura mater (a thick membrane that surrounds the brain and spinal cord) and the epidural space (space between the two layers of the dura mater).
47
Q

Do you know why lymphatic spread leads to haematogenous spread?

In which part of our body is the connection of the lymph to the blood?

A
  • Lymphatics drain into blood through the thoracic duct (left lymphatic duct) or the right lymphatic duct
48
Q

What determines the final destination of metastatic cells?

A
  • Blood flow characteristics and the structure of the vascular system can regulate the patterns of metastatic dissemination
  • Blood flow can favour certain organs as the metastasis targets, such is the case for breast and prostate cancer, which have predominantly bone metastases.
49
Q

Describe the route of prostate cancer spread

A
  • The figure illustrates the spread of prostate cancer cells via venous blood vessels (blue), lymph vessels (green), or nerves (yellow).
  • Once in the venous blood, CTCs are carried throughout the body via the blood supply, through arterial systems.
  • Once they have reached a suitable secondary site, like bone, cells extravasate from the blood and colonise the metastatic site.
50
Q

Give an examples of metastases that usually do not travel far distances to establish new tumours

A
  • In ovarian cancer, metastatic cells can grow in the peritoneal cavity, in the ascites fluid or by attaching to the surface of peritoneal organs
  • They do not establish metastases in other visceral organs.
51
Q

What are some tumour types that metastasise regardless of vascular anatomy or rate of blood flow

A
  • For example, in the case of melanomas, their metastases form largely in the lungs and also in pulmonary and ovarian tissue.
  • This highlights the important of “congenial soil”, or the metastatic niche.
52
Q

What is the metastatic niche?

A
  • a special microenvironment must be formed for metastatic cells to settle in their new location
53
Q

What are the four key stages that allow metastatic colonisation to occur?

A
  1. Priming
  2. Licensing
  3. Initiation
  4. Progression
54
Q

Describe priming

A
  • Highly proliferative cancer cells at the primary site become hypoxic (low oxygen levels) and inflammatory.
  • They release different systemic mediators including cytokines, ECM remodelling enzymes and extracellular vesicles (exosomes).
  • Exosomes are small, cell-derived vesicles that promote cell-cell communication.
  • They carry cargoes that can either accelerate of suppress metastasis.
  • These factors reach the pre-metastatic niche via the circulation and are believed to prepare it for subsequent invasion and thus prepare the formation of an immature pre-metastatic niche.
55
Q

Describe licensing

A
  • The ECM at the pre-metastatic niche is remodelled by recruitment of supporting cells, like bone marrow-derived cells and immune cells.
  • A diverse family of cytokines which regulate migration and homeostasis attract these supporting cells to the area.
  • Together with the host stroma, they remodel the local microenvironment and provide metastasis-promoting cytokines and chemokines.
  • A mature pre-metastatic niche is formed.
56
Q

Describe initation

A
  • Circulating cancer cells arrive at the new site and extensively remodel the new environment to promote their own growth
  • The ECM of the colonised site is broken down and new ECM components are produced by cancer cells, like matrix protein tenascin C produced by breast cancer cells in lungs.
  • At the same time, cancer cells send signals (like transforming growth factor (TGF)-β) to stromal cells, (e.g. fibroblasts) to deposit more matrix proteins, (e.g. periostin, a binding partner of tenascin C, or fibronectin)
  • Cancer cells also secrete crosslinking enzymes, like LOX (lysyl oxidase) and PLOD2 (Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase 2), to stiffen the ECM and increase integrin/focal adhesion signalling to favour metastasis.
  • As mentioned earlier, integrins are altered when cancer cells escape the primary tumour.
  • At the new site, integrins are altered again, this time to interact with the new extracellular matrix.
  • Only cells expressing integrins that facilitate tissue invasion are preferentially expressed.
  • Micrometastases are formed.
57
Q

Describe progression

A
  • The pre-metastatic niche can host more cancer cells and promote metastatic tumours to grow and expand.
  • A macrometastasis is now formed.
58
Q

How long does it take to get from primary tumour growth to metastatic colonisation?

A
  • The answer is not simple.
  • The metastatic process can occur early or late in primary tumour formation, with a timescale of a brief period or decades.
  • Furthermore, the metastatic process can pause at any point, leading to a state known as dormancy.
  • The dormant tumour cells can reside in new tissues or organs for many years, only to restart at some point to form a metastatic mass.
59
Q

What are disseminated tumour cells (DTCs)?

Describe them

A
  • circulating tumour cells (CTCs) which infiltrate distant organs and survive there are DTCs
  • They can be present for many years without developing a metastasis and have the ability to evade immune system recognition, are more resistant to chemodrugs and increase the likelihood of recurrence
60
Q

What is the dormant phase of metastatic cells?

A
  • Clinically, the dormant phase is described as the time between removal of the primary tumour and a relapse in a patient who was clinically disease free.
  • the metastatic process can be delayed by years due to the dormant phase
61
Q

What are 3 possible scenarios of how dormancy of metastatic cells come about?

they are not mutually exclusive

A
  1. Angiogenic dormancy:
    - During a period of impaired tumour angiogenesis, the size of the tumour is maintained by balancing cell proliferation and cell death.
  2. Immune-mediated dormancy
    - Cytotoxic immune-cells responses “trim” the number of proliferating tumour cells thus maintaining the cell death and proliferation balance.
  3. Cellular dormancy:
    - Solitary DTCs or DTC cell clusters (10-20 cells) enter a prolonged growth arrest with no increase in cell death
62
Q

Why is the dormant phase a promising therapeutic window?

What could be addressed?

A
  • it is a chance to target metastatic cells
  • but it s not an easy task as we do not have preclinical models that could be used to study the complexity of dormancy
  • there are several areas of interest that can be addressed, including:
  • identification of microenvironments that promote dormancy;
  • relationships between stem cell pathways and dormancy;
  • the molecules/triggers behind reactivation of dormant cells.
63
Q

What do in vitro models allow scientists to study?

A
  • they can study various cancer cell lines (cells derived from primary or metastatic tumours) in response to different treatments or conditions
64
Q

What can be observed by studying cancer cells in vitro?

A
  • how cancer cells migrate
  • e.g. towards different chemokines or organ-derived proteins
  • so the mechanism attracting cancer cells to different sites during metastatic processes can be made
65
Q

What assays and devices are used to study cancer cell migrations in in vitro studies?

A
  • Scratch assay (also known as wound-healing assay),
  • Boyden chambers
  • Micropatterning and pre-forming rings or paths
  • Microfluidic devices
66
Q

How can in vitro approaches also study the interactions between cancer cells and other cell types?

A
  • using co-culture studies
67
Q

Describe microfluidic devices

A
  • Microfluidic devices can be also used to study cancer cell interactions with other cells.
  • As illustrated below, a microfluidic device contains a set of micro-channels inside a mould (made of various polymers).
  • The micro-channels are connected together to be able to control fluid flow, but also to pump nutrients or drugs.
  • Using microfluidic devices, we can represent the blood flow, while also being able to co-culture different cell types.
68
Q

How do you recreate in vivo microenvironment in the microfluidic device?

A
69
Q

What are some in vivo models used to study human cancer?

A
  • amoebae
  • worms
  • flies
  • fish
  • mice
70
Q

How can we compare complex human organs and diseases, with simple organisms?

A
  • The key factor is that the fundamental biology that is conserved throughout
  • for example the genetic code that is conserved from bacteria to humans, or the eukaryotic cell structure
71
Q

Describe the use of social amoeba Dictyostelium discoideum

What was it used to study?

A
  • researchers designed a simple model to study directed cell migration and chemokine signalling
  • During chemotactic migration, amoeba acquire amoeboid migration to navigate complex environments at high speed.
  • The chemotactic migration is also observed in tumours
72
Q

What was Caenorhabditis elegans used to study?

Describe the study

A
  • to recreate the process of a cell crossing the basement membrane.
  • During the developmental stage of C.elegans, a single cell has to breach two underlying basement membranes as part of normal morphogenesis.
  • This simple model allowed scientists to recognise the first steps in activating the cell invasion programme.
  • Cell cycle is arrested in G1, chromatin is modified, and a set of transcription factors is upregulated.
  • In C.elegans one of the transcription factors was found to be FOS-1A, which is the orthologue of proto-oncogene FOS, a protein strongly associated with cancer cell invasion.
73
Q

What is orthotopic injection?

A
  • Cancer cells are injected into animals in different locations to represent different stages of cancer progression.
  • To represent the primary tumour, cancer cells are injected in the original site of the tumour (e.g. if cancer cells are from breast cancer they are injected into the breast tissue of the mouse).
  • This is called an orthotopic injection and it allows scientists to study the primary tumour growth and early metastatic events.
74
Q

What are xenograft models?

Why are they used instead of orthotropic injection?

A
  • However, as injecting into the original site of the tumour can be difficult, cancer cells are sometimes injected subcutaneously into the flank of the mouse where they can form tumours that can also used as models of the primary tumour.
  • These are usually referred to as xenograft models.
  • Strictly speaking, the orthotopic tumours are also xenografts since a xenograft is defined as tissue from one species being grafted into another species.
75
Q

What is intravenous injection used to study?

A
  • To study the circulation of cancer cells, they can also be injected into the blood vessels of mice.
  • This is called an intravenous injection and can help in studying metastatic colonisation.
  • Injecting cancer cells in locations other than the origin of the primary tumour benefits in the research of the final stage of colonisation and formation of metastasis.