Cancer Flashcards
(103 cards)
define tumour as an evolutionary proces
Tumourigenesis is an evolutionary process:
-Tumours arise and progress through Darwinian evolution within the body.
-Cells acquire random genetic mutations, and some of these provide selective advantages.
-These advantages may include faster proliferation, evasion of apoptosis, angiogenesis, and immune evasion.
Tumour cell populations are heterogeneous:
-As mutations accumulate, genetically distinct subclones arise within the tumour.
-This leads to intra-tumour heterogeneity, a key hallmark of cancer evolution.
Natural selection acts on cancer cells:
-Just like in ecological systems, cells that are more “fit” in a given environment will outcompete others.
-The tumour microenvironment (e.g., oxygen levels, immune surveillance) acts as a selective pressure.
Cancer evolution is both clonal and subclonal:
-Clonal evolution: Early mutations give rise to dominant clones.
-Subclonal evolution: Later mutations create branches of subpopulations, which coexist or compete.
Hallmarks of evolutionary tumour progression:
-Initiation: A single cell acquires a driver mutation.
-Promotion: Expansion of mutated clones.
-Progression: Accumulation of further mutations leads to metastasis and therapy resistance.
revise evolutionary concepts and apply them in the context of tumour progression
Mutation and genetic drift:
-Mutations occur randomly and can be neutral, deleterious, or advantageous.
-Some subclones expand not because they’re better but due to genetic drift in small populations.
Natural selection:
-Cells with beneficial mutations outgrow others (e.g., TP53 mutations for apoptosis resistance).
-Therapeutic pressure can also act as a selective force, promoting resistant clones.
Fitness landscapes:
-Different clones occupy different positions on a metaphorical “landscape” of fitness.
-Therapy or environmental changes reshape the fitness landscape, favouring new adaptive clones.
Convergent evolution:
-Different clones or patients may develop similar phenotypes (e.g., loss of PTEN), even via different genetic paths.
-Suggests that some evolutionary solutions are predictable.
Branching evolution:
-Most tumours do not evolve linearly.
-Instead, multiple lineages evolve in parallel, often visible through multi-region sequencing.
Neutral evolution:
-In some cases, subclones may expand without strong selection.
-These tumours evolve mainly by random drift, especially in early or low-grade stages.
To use selected examples to illustrate concepts based on this paper
Renal cell carcinoma (RCC):
-Ciriello discusses clear cell RCC, which often follows a branched evolutionary pattern.
-Early VHL mutations are followed by divergent pathways in different subclones.
-Shows how different regions of the same tumour have distinct mutational profiles.
Breast cancer (particularly ER+ subtypes):
-Often follows punctuated evolution—long periods of stasis interrupted by bursts of change.
-Therapy resistance often arises from pre-existing subclones or de novo mutations under treatment pressure.
Glioblastoma multiforme (GBM):
-Highly heterogeneous, with multiple coexisting clones showing convergent evolution (e.g., mutations in RTK pathways).
-Supports the concept that different routes can lead to the same functional tumour characteristics.
Chromothripsis:
-A phenomenon where massive genomic rearrangements occur in a single event.
-Illustrates how evolution in tumours can be saltatory (sudden) rather than gradual.
Therapy-induced evolution:
-Under chemotherapy or targeted therapy, subclones with resistance mutations (e.g., EGFR T790M in lung cancer) expand.
-shows how selection pressures drive tumour adaptation, much like antibiotic resistance in bacteria.
founder effect
a genetic phenomenon where a new population is established by a small group of individuals which may not carry all the genes of the initial larger population-resulting in decreased genetic diversity
what is adaptation
a heritable trait that has evolved through natural selection which enhances an individuals ability to survive and reproduce
Selection heterogeneity
-variation in the selective pressure experienced by different individuals or groups within a population, leading to different evolutionary outcomes for different parts of the population. This means that the same trait might be advantageous or disadvantageous depending on the specific environment or conditions an individual or group faces.
-In evolutionary biology, selection refers to the process where individuals with certain traits are more likely to survive and reproduce than others, leading to the spread of those traits within a population.
Type of cancer causing mutations
Driver mutations
- Genetic or epigenetic changes that directly contribute towards cancer development and progression
- Provide a growth or survival advantage, pushing it towards uncontrolled proliferation
- not only alter the function of the corresponding protein, but they do so in a way that promotes cancer-enabling features such as tumor initiation, progression, invasion, or resistance to therapy.
Passenger mutatuions
-Do not contribute toward cancer development progression
-Arise during the cell cycle as byproducts of genomic instability
-dont give cell any growth advantage
-mutational pattern observed in a gene or DNA region can be explained by neutral mutagenesis; hence
How driver and passenger mutations facilitate tumour progression
driver
- enable cancer hallmarks such as like sustained proliferation, resistance to cell death & invasion
- target for selection as cancer cells carrying driver aberrations are more likely to expand
Passenger
- increase tumour heterogeneity(increase genetic chaos)
- Passengers may interact indirectly with driver mutations as they create a genomic enviroment that support and enable future adaptations to occur(e.g-disable dna repair gene)
- large number of passenger mutations may be deleterious in bulk as they increase cell stress
Discuss how evolution in cancer differs from non cancerous population with applying the concept to genetic drift
-in non cancerous population genetic drift occurs which is the random fluctuation of of allele frequency over time due to mutations, leading to changes in the gene pool over time, this process is neutral & not driven by selective pressures
-However in cancerous cells, evolution is driven by genetic instability which leads to the accumulation of mutations.Mutations may be neutral or they may confer a selective advantage to these cells resulting in clonal expansion and tumour progression
-mutations that enhance survival and proliferation are positively selected for9aligning with neo darwinian theories)
-the tumor microenvironment (TME)—which includes immune cells, blood vessels, fibroblasts, etc.—interacts with cancer cells, applying selective pressures and promoting coevolution. That means cancer cells evolve in response to the TME, and vice versa (e.g., immune evasion or angiogenesis). In contrast, in non-cancerous populations, such intense local selection from the microenvironment is usually absent or much weaker, so coevolution isn’t a prominent evolutionary driver.
what are mutations and what are the different types of mutations
Mutations are the alterations in the nucleic acid sequence of a genome in an organism
-Genetic instability is often brought about due to cancers having defects in dna repair mechanisms leading to the accumulation of mutations
what are cancer stem cells
small subpopulation of cells within tumours with capabilities like(normal stem cells), self-renewal, differentiation and tumorigenicity
-Tumor initiation and progression
-Resistance to therapy (they often survive treatment that kills bulk tumor cells)
-Recurrence and metastasis
Cancer stem cells role in tumor evolution
-involed in tumor initiation & growth due to their self renewal capabilities and there ability to initiate tumor formation and drive continued expansion
-can survive treatment that would killdifferentiated cells due to their stem cell like properties
-can metastasize due to there plasticity and migratory abilites
-CSC give rise to intratumoral heterogeneity due to their ability to differentiate
Define and discuss intratumoral heterogenity
-ITH refers to the coexistance of many different cell types within the same tumor
-differences could be genetic,epigenetic,transcriptional or phenotypic
-Tumor cells exist in multiple states
- Tumor is not uniform, different regions & cells show different vulnerabilites
-ITH is dynamic and evolves over time due to pressures like treatment and immune surveillance
ITH is linked to:
poor clinical outcome, increased metastatic potential, greater chance of therapy resistance
-Cell states determine how cells respond to treatment, some cells may srvive & drive a relapse
Mechanismss by which cells can survive:
-Entering dormant state
-Due to epigenetic reprogramming & cellular plasticity
-Resistant subclones due to a combination of alterations that confer resistance
-TME interactions
-Activation of survival pathways due to stress
Definition of Genomic Instability:
Genomic instability refers to the increased tendency of the genome to acquire mutations and alterations over time. In cancer, this instability drives tumor heterogeneity and evolution, allowing cancer cells to adapt to selective pressures (e.g., therapy, immune surveillance). It is considered a hallmark of cancer because it enables the accumulation of the genetic diversity necessary for tumor progression and resistance.
Different Subtypes of Genomic Instability:
According to Ciriello’s paper, genomic instability in cancer can be broadly categorized into the following subtypes:
Single-Nucleotide Instability (SNI):
Characterized by point mutations or small insertions/deletions (indels).
Common in cancers with defects in DNA repair pathways, especially mismatch repair (MMR).
Seen in tumors with microsatellite instability (MSI).
Chromosomal Instability (CIN):
Involves gains, losses, or rearrangements of whole chromosomes or large segments.
Results in aneuploidy (abnormal number of chromosomes) and structural rearrangements.
Can be due to errors in mitosis, centrosome amplification, or defects in the spindle checkpoint.
Genome Doubling:
A specific form of instability where the entire genome is duplicated, leading to tetraploidy.
Often a precursor to chromosomal instability, as tetraploid cells are more prone to segregation errors.
Kataegis and Mutational Clusters:
Localized hypermutation phenomena, typically due to the activity of APOBEC cytidine deaminases.
Leads to mutation clusters in specific genomic regions.
Structural Variants and Rearrangement Instability:
Involves complex chromosomal rearrangements, such as chromothripsis (shattering and reassembly of chromosomes).
These dramatic events can create oncogenic fusions or disrupt tumor suppressor genes.
The Impact of Therapy on Tumour Evolution
The Role of Intratumour Heterogeneity (ITH) in Therapy Response
💊 ITH and Therapy Response:
1. Selective Pressure from Treatment:
Therapy kills sensitive subclones, but spares resistant ones, which expand and dominate after treatment.
This clonal selection drives tumour evolution and drug resistance
📍 Example:
Subclones that have lost the TP53 tumour suppressor gene can evade apoptosis induced by chemotherapy.
TP53-mutant cells often survive treatment and seed relapse.
- Pre-existing vs Acquired Resistance:
Pre-existing resistant subclones may be present at low levels before treatment.
Upon therapy, these subclones become dominant.
Resistance can also arise de novo through new mutations under therapeutic pressure.
📍 Example:
In melanoma, BRAF inhibitor therapy may initially work, but subclones with NRAS mutations or MEK pathway reactivation expand, driving resistance. - Phenotypic Plasticity:
Some resistant subclones exhibit non-genetic resistance via cell state changes, not DNA mutations.
📍 SOX10 Example (from the paper):
In melanoma, therapy-resistant cells downregulate SOX10, a transcription factor critical for the melanocyte lineage.
This leads to reprogramming into a neural crest–like, drug-tolerant state, allowing the cells to survive therapy without needing genetic mutations.
This plasticity contributes to minimal residual disease and relapse. - Spatial and Temporal Heterogeneity:
Different regions of the tumour (or metastases) may harbour different subclones, leading to partial treatment response.
Over time, the tumour evolves under therapy, making early biopsies insufficient to guide later treatment. - Immune Evasion and Immunotherapy:
ITH can cause loss of clonal neoantigens, allowing resistant subclones to escape immune detection.
📍 Example:
A tumour might lose HLA expression or mutate antigen-presentation machinery in certain clones, evading cytotoxic T cells.
⚠️ Clinical Implications:
High ITH = poor prognosis, higher relapse rate, and reduced response to mono-therapies.
Calls for:
Multi-targeted therapy
Adaptive therapy (changing drugs as clones evolve)
Monitoring clonal dynamics using liquid biopsy or single-cell sequencing
Definition of cancer
Definition Overview:
-Cancer is defined not as a single disease, but as a group of diseases driven by acquired genetic and epigenetic alterations that enable cells to override normal regulatory mechanisms and evolve progressively through stages of initiation, promotion, and progression.
-These changes result in cells acquiring distinct capabilities – known as the “hallmarks of cancer” – that allow them to survive, proliferate, and spread uncontrollably.
🧩 Cancer as a Disease of Genetic and Epigenetic Evolution
-Fouad emphasizes that cancer arises due to the accumulation of mutations in oncogenes, tumour suppressor genes, and DNA repair genes.
-Additionally, epigenetic reprogramming and tumour microenvironment (TME) interactions shape how these mutations manifest phenotypically.
🔑 Core Characteristics (Hallmarks):
Below are the hallmarks of cancer as revisited in the paper, each with detailed explanation and real biological examples to deepen understanding.
- Sustaining Proliferative Signaling
Cancer cells bypass normal growth regulation and continuously signal themselves (or others) to divide.
Example: Activation of RAS oncogene → continuous MAPK pathway signaling → uncontrolled proliferation.
Mechanism: Mutant RAS is GTP-bound permanently, triggering constant downstream signaling without needing a growth factor. - Evading Growth Suppressors
Cancer cells inactivate pathways that would normally halt cell division.
Example: Loss-of-function mutations in TP53 or RB1 (tumour suppressors).
TP53 = the “guardian of the genome” – its loss allows damaged cells to continue dividing.
RB1 loss = inability to restrict E2F → uncontrolled entry into S-phase of the cell cycle. - Resisting Cell Death
Cancer cells evade programmed cell death (apoptosis), allowing them to survive despite major damage or stress.
Example: Overexpression of BCL-2 in B-cell lymphomas blocks mitochondrial apoptosis.
Loss of p53 also prevents apoptosis in response to DNA damage. - Enabling Replicative Immortality
Tumour cells bypass replicative senescence and divide indefinitely.
Example: Reactivation of telomerase (TERT) in 90% of cancers.
Normal cells stop dividing when telomeres become critically short; cancer cells maintain telomere length, avoiding senescence. - Inducing Angiogenesis
Tumours stimulate the formation of new blood vessels to ensure adequate supply of oxygen and nutrients.
Example: Upregulation of VEGF in response to hypoxia.
Tumour hypoxia → stabilization of HIF-1α → VEGF secretion → new capillaries grow toward tumour. - Activating Invasion and Metastasis
Cancer cells acquire the ability to break away from the primary tumour and colonize distant organs.
Example: Loss of E-cadherin (adhesion molecule) in epithelial cancers.
EMT (epithelial-to-mesenchymal transition) programs are activated – involving TWIST, SNAIL, and ZEB – allowing cells to become migratory and invasive. - Deregulating Cellular Energetics
Cancer cells reprogram their metabolism to favor glycolysis, even in the presence of oxygen.
Example: The Warburg effect – upregulation of GLUT1, HK2, and LDHA to promote aerobic glycolysis.
Supports rapid ATP generation and biosynthesis needed for growth. - Avoiding Immune Destruction
Tumours evolve mechanisms to evade recognition and elimination by the immune system.
Example: Upregulation of PD-L1 in tumour cells binds PD-1 on T cells → T cell exhaustion.
Loss of MHC I expression prevents cytotoxic T cell recognition. - Genome Instability and Mutation
Cancer cells exhibit increased mutation rates due to defective DNA repair.
Example: Mutations in BRCA1/2 (homologous recombination repair genes) lead to double-strand break accumulation.
Also includes chromothripsis and microsatellite instability (MSI) in mismatch repair-deficient tumours. - Tumour-Promoting Inflammation
Chronic inflammation provides growth factors, cytokines, and mutagenic ROS that fuel tumour growth.
Example: In colorectal cancer, chronic inflammation from IBD promotes IL-6 and TNF-α secretion, stimulating tumourigenesis.
Macrophages in the TME may secrete MMPs that degrade ECM and promote invasion.
Additional Context from Paper:
Cancer is a multifaceted evolutionary process, not a linear progression.
Subclonal evolution is key to understanding treatment resistance.
Plasticity is emphasized, where cells switch phenotypes (e.g., SOX10 loss in melanoma leading to therapy-resistant dedifferentiated states, similar to what Ciriello describes).
Final Definition Summary (In Fouad’s View):
Cancer is a complex, dynamic, and adaptive system where cells evolve to acquire multiple capabilities (“hallmarks”) that override normal control mechanisms. These changes are driven by a combination of genetic instability, clonal selection, and interactions with the tumour microenvironment, enabling survival, growth, and dissemination.
List the hallmarks of cancer as stated in the article and give a brief description of each
- Selective Growth and Proliferative Advantage
Cancer cells acquire the ability to grow and divide autonomously, ignoring normal growth-regulatory signals.
This includes activation of oncogenes (e.g. RAS, MYC) and inactivation of tumour suppressor genes (e.g. TP53, RB1).
Cancer cells sustain chronic proliferative signaling through mutations or autocrine loops, giving them a growth advantage over normal cells. - Altered Stress Response Favoring Overall Survival
Cancer cells develop mechanisms to withstand various types of cellular stress, such as DNA damage, oncogene activation, or metabolic stress.
A key feature is the evasion of apoptosis, often via loss of p53 or upregulation of anti-apoptotic proteins (e.g. BCL-2).
These cells can survive under conditions that would kill normal cells, allowing malignant progression. - Vascularization
Tumours require angiogenesis to grow beyond a minimal size and to metastasize.
Cancer cells upregulate angiogenic factors such as VEGF and manipulate endothelial cells to form new blood vessels.
Tumour-induced angiogenesis is often abnormal, resulting in leaky, disorganized vasculature. - Invasion and Metastasis
Cancer cells acquire the ability to detach, migrate, invade surrounding tissues, and colonize distant organs.
This involves epithelial-to-mesenchymal transition (EMT), loss of cell adhesion molecules like E-cadherin, and extracellular matrix degradation by enzymes like MMPs.
Metastasis is the primary cause of cancer-related death. - Metabolic Rewiring
Cancer cells reprogram their metabolism to support uncontrolled growth and survival.
They rely more on aerobic glycolysis (Warburg effect) rather than oxidative phosphorylation.
This shift supports rapid ATP production and biosynthesis of macromolecules needed for proliferation. - An Abetting Microenvironment
The tumour microenvironment (TME), including stromal cells, fibroblasts, and immune cells, actively supports tumour development.
Cancer cells co-opt surrounding non-malignant cells to promote growth, angiogenesis, immune evasion, and metastasis.
Fibroblasts, for example, become cancer-associated fibroblasts (CAFs), which secrete growth factors, cytokines, and ECM-degrading enzymes. - Immune Modulation
Tumours develop mechanisms to avoid immune recognition and destruction, and may even exploit immune cells to support their own growth.
This includes downregulation of antigen presentation, expression of immune checkpoint ligands (e.g. PD-L1), and secretion of immunosuppressive cytokines.
Tumour-associated macrophages (TAMs) and regulatory T cells (Tregs) create an immunosuppressive TME.
Q: How do benign and malignant growths differ in their cellular characteristics and behaviors?
A:
Benign growths consist of well‑differentiated cells that closely resemble their tissue of origin, proliferate slowly, and expand by pushing adjacent structures aside without invading them. They remain encapsulated, do not breach the basement membrane, and never metastasize. In contrast, malignant growths are composed of poorly differentiated or undifferentiated cells that proliferate rapidly and infiltrate surrounding tissues. They acquire the ability to degrade extracellular matrix via matrix metalloproteinases, invade stroma, enter blood or lymphatic vessels, and establish new colonies at distant sites (metastasis). These invasive and metastatic properties are absent in benign lesions.
What “tips the scale” from a benign lesion toward malignancy, and which hallmarks are involved in that transition?
A:
The critical shift occurs when cells acquire capabilities for invasion & metastasis, vascularization, and immune modulation. A benign lesion may already exhibit selective growth advantage, altered stress response, metabolic rewiring, and an abetting microenvironment, allowing it to expand locally. However, without (1) the ability to invade surrounding tissues and enter vasculature (Hallmark 4), (2) the recruitment of new, often disorganized blood vessels to feed and facilitate intravasation (Hallmark 3), and (3) mechanisms to evade or co‑opt immune surveillance (Hallmark 7), the lesion cannot disseminate. Once one or more of these malignant hallmarks is gained—often through additional genetic or epigenetic hits—the lesion tips into true malignancy.
Which of Fouad’s seven hallmarks are common to both benign and malignant lesions, and why aren’t they sufficient for metastasis on their own?
A:
Both benign and malignant growths share these four core capabilities:
Selective Growth & Proliferative Advantage – mutations (e.g. activated RAS, MYC overexpression) or loss of TP53/RB1 allow sustained division.
Altered Stress Response Favoring Survival – evasion of apoptosis via p53 loss or BCL‑2 upregulation lets cells endure DNA damage or oncogenic stress.
Metabolic Rewiring – adoption of the Warburg effect (aerobic glycolysis) and increased nutrient uptake supports biomass accumulation.
An Abetting Microenvironment – stromal cells, CAFs, and secreted factors (e.g. TGF‑β, cytokines) fuel growth and survival.
what is carcinogenesis
Carcinogenesis (also called oncogenesis or tumorigenesis) is the process by which normal cells are transformed into cancer cells, involving progressive accumulation of mutations and epimutations that disrupt normal regulatory circuits
What is the cell cycle? Is this process regulated? If so, how?
The cell cycle is the tightly controlled process by which a cell duplicates its contents and divides. It consists of four major phases: G1, S, G2, and M.
Yes, the cell cycle is highly regulated by a network of checkpoint proteins and tumor suppressors. For example:
-Cyclin-dependent kinases (CDKs) and cyclins drive progression from one phase to the next.
-RB (retinoblastoma protein) is a gatekeeper at the G1/S checkpoint—it prevents replication unless conditions are favorable.
-p53 halts the cycle in response to DNA damage, allowing repair or triggering apoptosis.
In cancer, mutations in regulators like RB and p53 lead to loss of control, enabling cells to proliferate even when DNA is damaged.
What is meant by growth signals?
Growth signals refer to external or internal molecular cues that stimulate cells to exit quiescence (G0) and re-enter the cell cycle, leading to proliferation. In normal tissue, these are tightly regulated, but in cancer, these signals become hyperactive, constitutive, or ligand-independent, fueling uncontrolled division.