Neoplasia & Immunodeficiency Syndrome Flashcards

1
Q

Define Tumor and neoplasm

A

Tumor: Abnormal swelling, which can be benign (non-cancerous) or malignant (cancerous).

Neoplasm: An abnormal mass of tissue with excessive and uncoordinated growth that persists even after the initial trigger is removed.

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

What are the features of neoplasm and Neuplasia and Cancer

A

Key Features of Neoplasms

Purposeless Growth: The growth serves no functional purpose.

Progressive Growth: The tumor continues to grow over time.

Parasitic Growth: The growth disregards the body’s needs and consumes resources.

Aberrant Cell Growth: Imbalance between cell division (mitosis) and cell death (apoptosis).

Clonal Evolution: Tumor cells evolve through mutations.

Differentiation (Grading): The extent to which tumor cells resemble normal cells.

Neoplasia

The process of tumor formation.

Cancer

Refers to malignant tumors that invade surrounding tissues and can spread to distant sites (metastasis).

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

What’s Teratoma?

A

A teratoma is a neoplasm arising from pluripotent germ cells that can contain a diverse range of tissue types derived from more than one of the three germ layers: ectoderm, mesoderm, and endoderm.

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

What are the types of tumor?

A

Types of Tumors

Hamartoma: Non-cancerous, disorganized growth of native tissue cells.

Choristoma: Non-cancerous growth of normal cells in an abnormal location.

Difference Between Hamartoma and Choristoma:

Hamartoma: Disorganized growth in the original tissue.

Choristoma: Normal cells in an ectopic (wrong) site.

Teratoma:

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

What are the hallmark of cancer

A

Self-Sufficiency in Growth Signals

Cancer cells can grow without the need for external signals that normal cells require for growth.

Example: Mutations in growth factor receptors can cause continuous activation, leading to uncontrolled cell proliferation.

Insensitivity to Growth-Inhibitory Signals

Cancer cells ignore signals that normally inhibit cell growth.

Example: Mutations in tumor suppressor genes like RB or TP53 remove these inhibitory controls.

Altered Cellular Metabolism

Cancer cells often shift their energy production to aerobic glycolysis, known as the Warburg effect, to support rapid growth.

Example: Increased glucose uptake and lactate production even in the presence of oxygen.

Evasion of Apoptosis

Cancer cells avoid programmed cell death, allowing them to survive longer than normal cells.

Example: Mutations in genes regulating apoptosis, like BCL-2, lead to resistance to cell death.

Limitless Replicative Potential (Immortality)

Cancer cells can divide indefinitely, unlike normal cells that have a limited number of divisions.

Example: Activation of telomerase enzyme maintains telomere length, allowing endless replication.

Sustained Angiogenesis

Cancer cells stimulate the growth of new blood vessels to supply the tumor with nutrients and oxygen.

Example: Overexpression of VEGF (vascular endothelial growth factor) promotes blood vessel formation.

Invasion and Metastasis

Cancer cells can invade surrounding tissues and spread to distant sites in the body.

Example: Changes in adhesion molecules like E-cadherin and increased activity of enzymes like matrix metalloproteinases help in breaking through tissue barriers.

Evasion of Immune Surveillance

Cancer cells develop mechanisms to avoid detection and destruction by the immune system.

Example: Expression of proteins that inhibit immune responses, like PD-L1, helps cancer cells evade immune attacks.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

List the Examples of Cancer Genes

A

RB Gene and RB Protein: RB gene mutations disrupt cell cycle control, promoting uncontrolled cell growth.

TP53 and p53: TP53 gene mutations prevent the p53 protein from inducing apoptosis, aiding in cell survival.

MYC: MYC gene amplification leads to increased cell proliferation and growth.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What are the characteristics to enable or enhance this hallmark

A

Cancer-Promoting Inflammation

Chronic inflammation can lead to a microenvironment that supports cancer development and progression.

Genomic Instability

An increased rate of mutations in cancer cells creates genetic diversity, allowing them to adapt and evolve.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Explain how cancerous cells become Self-Sufficiency in Growth Signals

A

Cancer cells become self-sufficient in growth signals through mutations that enable them to bypass normal regulatory mechanisms. These mutations can occur at various points in the growth signaling pathway, including growth factors, growth factor receptors, and downstream signaling proteins

Growth Factors: Autocrine Stimulation:

Some cancer cells produce growth factors to which they themselves are responsive.
Paracrine Stimulation:

Tumor cells can induce surrounding stromal cells to produce growth factors that support tumor growth.

Growth Factor Receptors
Growth factor receptors can become oncogenic through various mutations or overexpression.

Overexpression:Mutations and Gene Rearrangements:

These can cause receptors to become constitutively active, meaning they send growth signals even without growth factors.

Downstream Signal-Transducing Proteins
Mutations in proteins that transmit signals from growth factor receptors to the nucleus can also drive cancer growth.

RAS Protein:

RAS mutations lead to continuous signal transmission for cell growth.

ABL Protein:

ABL mutations can lead to uncontrolled growth signals

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are Oncoproteins, Oncogenes, Proto-Oncogenes

A

Proto-Oncogenes: These are normal cellular genes that promote cell proliferation under controlled conditions.

Oncogenes: These are mutated or overexpressed versions of proto-oncogenes that function independently of normal growth-promoting signals, driving increased cell proliferation.

Oncoproteins: Proteins encoded by oncogenes that drive cancer cell proliferation. These proteins may arise from various genetic aberrations.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What are the Key Mutations Leading to Sustained Proliferation

A

Growth Factor Receptors

EGF Receptor Tyrosine Kinase Activation: Point mutations in EGF receptor can activate this kinase (e.g., lung cancer).

HER2 Receptor Tyrosine Kinase Activation: Gene amplification of HER2 receptor leads to its activation (e.g., breast cancer).

JAK2 Tyrosine Kinase Activation: Point mutations in JAK2 kinase (e.g., myeloproliferative neoplasms).

Nonreceptor Tyrosine Kinases

ABL Activation: Chromosomal translocation creates a BCR-ABL fusion gene (e.g., chronic myeloid leukemia, acute lymphoblastic leukemia).

Downstream Signaling Molecules

RAS Activation: Point mutations in RAS lead to its activation (e.g., many cancers).

PI3K and BRAF Activation: Point mutations activate these serine/threonine kinases (e.g., many cancers).

Transcription Factors

MYC Expression: MYC, a master transcription factor, is increased by chromosomal translocations (e.g., Burkitt lymphoma), gene amplification (e.g., neuroblastoma), and increased activity of upstream signaling pathways (e.g., many cancers).

Cell Cycle Regulators

CDK4/D Cyclin Complexes: Mutations that increase the activity of these complexes promote cell cycle progression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What’s the Mechanisms of Aberrant Signaling in cancer?

A

Mechanisms of Aberrant Signaling

Point Mutations: Alter specific amino acids in proteins, leading to their constant activation.

Gene Amplifications: Increase the number of copies of a gene, leading to overexpression of its protein product.

Chromosomal Translocations: Chromosomal translocations occur when parts of two different chromosomes break off and swap places. This can create fusion genes that produce hybrid proteins with new, often harmful functions.

Increased Expression: Enhanced transcription (copying of DNA to RNA) or stability of oncogenes can lead to overproduction of oncoproteins

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Examples of Oncoproteins and Their Mechanisms

A

Examples of Oncoproteins and Their Mechanisms

EGF Receptor Tyrosine Kinase: Point mutations activate this receptor in lung cancer.

HER2 Receptor Tyrosine Kinase: Gene amplification activates this receptor in breast cancer.

JAK2 Tyrosine Kinase: Point mutations activate this kinase in myeloproliferative neoplasms.

BCR-ABL Fusion Protein: Created by chromosomal translocation in chronic myeloid leukemia.

RAS Protein: Point mutations activate RAS in various cancers.

PI3K and BRAF Kinases: Point mutations activate these kinases in many cancers.

MYC Transcription Factor: Overexpressed due to translocations, amplifications, or increased upstream signaling.

CDK4/D Cyclin Complexes: Mutations increase the activity of these complexes, promoting cell cycle progression.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Cancer cells sustain proliferative signaling through mutations in proto-oncogenes, converting them into oncogenes that produce oncoproteins. These oncoproteins drive continuous cell proliferation by various mechanisms, including point mutations, gene amplifications, chromosomal translocations, and increased gene expression.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What are the Two primary tumor suppressor genes

A

Rb gene and the p53 gene.
These genes encode the retinoblastoma (RB) protein and the p53 protein, respectively,

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Whats called the Governor of the Cell Cycle?

A

RB

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

The RB protein is crucial for controlling?

A

the transition from the G1 phase to the S phase of the cell cycle, where DNA replication occurs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How do cancer cells evade growth suppressors?
In RB

A

Cancer cells can disrupt the normal regulatory function of RB through several mechanisms, leading to uncontrolled cell proliferation.

Loss-of-Function RB Mutations

Mutations in the RB gene can result in the production of a nonfunctional RB protein that cannot bind to E2F transcription factors. Without functional RB, there is no inhibition of E2F, and the cell cycle progresses unchecked, contributing to tumor development.

RB Mutation: Nonfunctional RB → E2F is not inhibited → Uncontrolled cell cycle progression.

CDK4 and Cyclin D Amplifications

The genes encoding CDK4 and cyclin D can be amplified or overexpressed in cancer cells. This leads to an increased formation of CDK4/cyclin D complexes, resulting in excessive phosphorylation and inactivation of RB. Consequently, E2F is continually released, promoting unregulated cell division.

CDK4/Cyclin D Overexpression: Increased RB phosphorylation → Inactivation of RB → Continuous release of E2F → Unchecked cell cycle progression.

Loss-of-Function Mutations in CDK Inhibitors

CDK inhibitors like p16/INK4a normally inhibit the activity of CDK4 and CDK6, preventing them from phosphorylating RB. Loss-of-function mutations in CDK inhibitors remove this inhibitory control, leading to increased RB phosphorylation and inactivation. This again results in the release of E2F and unregulated cell cycle progression.

CDK Inhibitor Mutation: Reduced inhibition of CDK4/6 → Increased RB phosphorylation → Inactivation of RB → Continuous release of E2F → Uncontrolled cell cycle progression.

Viral Oncoproteins

Certain viruses produce oncoproteins that can directly bind to and inactivate RB. For example, the E7 protein of human papillomavirus (HPV) binds to hypophosphorylated RB, preventing it from inhibiting E2F. This leads to the release of E2F and promotes uncontrolled cell division, contributing to the development of cancers such as cervical cancer.

Viral Oncoprotein (e.g., HPV E7): Binds hypophosphorylated RB → Prevents RB from inhibiting E2F → Continuous release of E2F → Unregulated cell cycle progression

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Mechanism of RB Action
Hypophosphorylated RB
In its hypophosphorylated (low-phosphate) state, RB is active and performs its role as a tumor suppressor by binding to E2F transcription factors. E2F transcription factors are essential for the transcription of genes needed for DNA synthesis. When RB binds to E2F, it inhibits E2F’s ability to promote the transcription of these genes, thereby preventing the cell from proceeding to the S phase of the cell cycle, where DNA replication occurs. Essentially, hypophosphorylated RB acts as a brake on cell cycle progression at the G1/S checkpoint.

Active RB: Binds E2F → Inhibits transcription of DNA synthesis genes → Cell cycle arrest at G1/S checkpoint.
Hyperphosphorylated RB
When cells receive growth signals through growth factor receptors, these signals activate cyclin-dependent kinases (CDKs), particularly CDK4 and CDK6, which partner with cyclin D. These CDK-cyclin complexes phosphorylate RB. The phosphorylation changes RB’s conformation, making it unable to bind to E2F transcription factors. As a result, E2F is released and can activate the transcription of genes required for DNA synthesis, allowing the cell to enter the S phase and proceed with cell division.

Inactivated RB (hyperphosphorylated): Cannot bind E2F → E2F free to promote gene transcription for DNA synthesis → Cell cycle progression to S phase.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How do cancer cells evade growth suppressors?
In p53

A

Cancer cells often circumvent the tumor-suppressive actions of p53 through various mechanisms, leading to uncontrolled cell growth and survival.

Loss-of-Function Mutations

The majority of human cancers show biallelic loss-of-function mutations in the p53 gene. This means both copies of the gene are mutated, rendering the p53 protein nonfunctional. Without functional p53, cells lose the ability to undergo cell cycle arrest or apoptosis in response to DNA damage, allowing mutations to accumulate and contribute to cancer progression.

Loss-of-Function Mutations: Mutations in both copies of the p53 gene → Nonfunctional p53 → Failure to induce cell cycle arrest or apoptosis → Accumulation of genetic damage.

Li-Fraumeni Syndrome

Li-Fraumeni Syndrome is a hereditary disorder where patients inherit one defective copy of the p53 gene. These individuals have a very high risk of developing various types of cancers because the remaining normal allele is highly susceptible to somatic mutation, leading to a complete loss of p53 function.

Li-Fraumeni Syndrome: Inherited defective p53 allele → High risk of cancer due to loss of the second allele.

Viral Oncoproteins

Certain viruses produce oncoproteins that can bind to and degrade p53, effectively neutralizing its tumor suppressor functions. For instance, the E6 protein from human papillomavirus (HPV) binds to p53 and promotes its degradation, thereby preventing p53 from inducing cell cycle arrest or apoptosis.

Viral Oncoproteins: HPV E6 protein binds to p53 → Degradation of p53 → Loss of tumor suppressor function → Increased risk of cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Mechanism of p53 Action
Activation by Stress Signals
p53 is a critical protein that responds to various cellular stress signals, such as DNA damage, low oxygen levels (hypoxia), and the activation of oncogenes. When cells experience stress, kinases such as ATM (ataxia-telangiectasia mutated) and ATR (ATM and Rad3-related) are activated. These kinases phosphorylate p53, which stabilizes and activates it by preventing its degradation. Normally, p53 is kept at low levels in the cell through the action of the protein MDM2, which tags p53 for degradation. Phosphorylation of p53 by ATM/ATR inhibits MDM2’s ability to bind p53, thus protecting it from degradation.

Stress Signal Activation: DNA damage activates ATM/ATR kinases → Phosphorylation of p53 → Inhibition of MDM2-mediated degradation → Stabilization and activation of p53.
Cell Cycle Arrest and DNA Repair
Once activated, p53 induces the expression of several target genes, including p21, a cyclin-dependent kinase inhibitor. p21 binds to and inhibits CDKs, leading to cell cycle arrest at the G1/S checkpoint. This pause allows the cell time to repair DNA damage before proceeding with division. By halting the cell cycle, p53 ensures that damaged DNA is not replicated and passed on to daughter cells.

Cell Cycle Arrest: Activated p53 induces p21 expression → p21 inhibits CDKs → Cell cycle arrest at G1/S checkpoint → DNA repair.
Induction of Senescence or Apoptosis
If the DNA damage is too severe to be repaired, p53 can initiate cellular senescence or apoptosis. Senescence is a state of permanent cell cycle arrest, while apoptosis is programmed cell death. By driving damaged cells into senescence or apoptosis, p53 prevents them from continuing to divide and potentially forming tumors.

Senescence or Apoptosis: Irreparable DNA damage → p53 triggers senescence or apoptosis → Prevention of propagation of damaged cells.

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

List other tumor suppressing genes

A

Other Tumor Suppressor Genes

In addition to RB and p53, several other tumor suppressor genes play significant roles in preventing uncontrolled cell growth:

APC (Adenomatous Polyposis Coli): A key regulator of WNT signaling, often mutated in colorectal cancers.

E-cadherin: Involved in cell-cell adhesion; its loss is associated with increased tumor invasiveness.

CDKN2A: Encodes p16/INK4a, a CDK inhibitor; its loss leads to unchecked cell cycle progression.

PTEN: A phosphatase that antagonizes PI3K/AKT signaling, often mutated in various cancers.

VHL (Von Hippel-Lindau): Regulates hypoxia-inducible factors (HIFs); mutations lead to renal cell carcinoma and other cancers.

STK11 (LKB1): Involved in cell polarity and energy homeostasis; mutations are linked to Peutz-Jeghers syndrome and various cancers.

TGF-β Pathway: Involved in growth inhibition and differentiation; mutations or disruptions in this pathway can lead to tumor progression.

22
Q
  1. Resisting cell death
    Cancer cells are able to evade apoptosis by acquiring anti-apoptotic regulators as follows:
    loss of P53 tumor suppressor function, which eliminates this critical damage sensor from the apoptosis-inducing circuit.
    Increasing the expression of antiapoptotic regulators (Bcl-2, Bcl-XL Mcl- 1).
    By downregulating proapoptotic Bcl-2–related factors (Bax,Bim,Apaf-1 ).
    Lesions that incapacitate the intrinsic (mitochondrial) pathway of apoptosis appear to be most common in cancers.
    In greater than 85% of follicular B-cell lymphomas, the antiapoptotic gene BCL2 is overexpressed due to a (14;18) translocation.
    Overexpression of other BCL2 family members is also linked to cancer cell survival and drug resistance
  2. Limitless replicative potential
A
23
Q

What are the ways cancer cells resist cell death?

A

Loss of p53 Function

Mechanism: The tumor suppressor protein p53 is crucial for detecting cellular stress and damage, such as DNA damage or hypoxia (low oxygen levels). When p53 detects such stress, it can initiate apoptosis to prevent damaged cells from surviving and proliferating.

Disruption in Cancer: Many cancers have mutations that inactivate p53. Without functional p53, the cell loses a critical sensor and mediator of apoptosis. This allows cells with damaged DNA to survive, continue dividing, and accumulate further mutations, contributing to cancer progression.

  1. Increased Expression of Antiapoptotic Regulators

Mechanism: Proteins like Bcl-2, Bcl-XL, and Mcl-1 prevent apoptosis by inhibiting the release of proapoptotic factors from the mitochondria, a key step in the intrinsic pathway of apoptosis. These antiapoptotic proteins help cells avoid self-destruction.

Disruption in Cancer: Cancer cells often produce higher levels of these antiapoptotic proteins, blocking apoptosis and thus promoting cell survival. This makes cancer cells more resistant to treatments that would typically induce cell death.

  1. Downregulation of Proapoptotic Factors

Mechanism: Proapoptotic proteins such as Bax, Bim, and Apaf-1 facilitate apoptosis by promoting the release of cytochrome c from the mitochondria, which activates caspases (enzymes that execute apoptosis).

Disruption in Cancer: Many cancer cells decrease the levels of these proapoptotic proteins, weakening the apoptotic response. This allows cancer cells to avoid programmed cell death and continue growing.

  1. Lesions in the Intrinsic (Mitochondrial) Pathway

Mechanism: The intrinsic pathway of apoptosis is controlled by signals from the mitochondria, which depend on a balance between proapoptotic and antiapoptotic proteins. This balance determines whether a cell will undergo apoptosis.

Disruption in Cancer: Mutations or alterations in genes that regulate this pathway are common in cancer. These changes can prevent the initiation of apoptosis, allowing cancer cells to survive despite signals that would normally induce cell death.

Example: Follicular B-Cell Lymphomas

Mechanism: In over 85% of follicular B-cell lymphomas, the gene BCL2, which encodes an antiapoptotic protein, is overexpressed due to a specific chromosomal translocation (t(14;18)).

Consequence: This overexpression of BCL2 leads to increased survival of these cancer cells. It makes them resistant to apoptosis and contributes to their resistance to cancer therapies, which often work by inducing apoptosis in cancer cells.

24
Q

Telomere Shortening in Normal Cells
Mechanism: During each cell division, a small portion of the telomere is lost. When telomeres become critically short, cells either enter a state of senescence (a p53-dependent arrest) or undergo apoptosis.
Consequence: This acts as a natural barrier to uncontrolled cell proliferation.

A
25
Q

How does cancer cells achieve Limitless Replicative Potential?

A

Telomerase Activation in Cancer Cells

Mechanism: Telomerase is an enzyme that adds repetitive nucleotide sequences to the ends of telomeres, effectively maintaining their length.

Disruption in Cancer: Many cancer cells reactivate telomerase, allowing them to maintain their telomeres and continue dividing indefinitely.

Consequence: This enables cancer cells to acquire a stem cell-like property, giving them the potential for limitless replication and contributing to tumor growth and progression.

26
Q

How do cancer cells induce Angiogenesis

A

Disruption in Cancer

VEGF Overexpression: Many cancers overexpress VEGF, leading to excessive angiogenesis. This supports tumor growth by supplying nutrients and oxygen.

27
Q

VEGF Inhibitors: Drugs that inhibit VEGF are used to treat advanced cancers. While these inhibitors can prolong the clinical course of the disease, they are not curative.

A
28
Q

Mechanism of Angiogenesis
Hypoxia-Induced Factor 1α (HIF1α): Hypoxia, or low oxygen levels, in the tumor environment triggers the activation of HIF1α. This transcription factor increases the expression of Vascular Endothelial Growth Factor (VEGF), a key pro-angiogenic molecule that stimulates the formation of new blood vessels.
Other Regulatory Factors: Various other molecules also regulate angiogenesis. For example:
p53: This tumor suppressor gene induces the synthesis of thrombospondin-1, an inhibitor of angiogenesis.
RAS, MYC, and MAPK Signaling: These pathways enhance the expression of VEGF, promoting angiogenesis and thereby facilitating tumor growth.

A
29
Q

List the Mechanism of Invasion in cancer cells

A

Loosening of Cell-Cell Contacts: Cancer cells lose their tight adhesion to each other by inactivating E-cadherin, a protein crucial for cell-cell adhesion.

Degradation of the Extracellular Matrix (ECM):

Attachment to New ECM Components: Cancer cells attach to novel ECM components exposed by the degradation process.

Migration of Tumor Cells: The cells migrate through the degraded ECM towards blood vessels or lymphatics.

30
Q

Tumor cells and stromal cells secrete proteolytic enzymes, such as _____&____ that degrade the basement membrane and interstitial matrix.

A

Matrix Metalloproteinases (MMPs) and cathepsins

31
Q

Some cancers preferentially metastasize to specific organs due to?

A

due to the expression of adhesion molecules or chemokine receptors that match ligands expressed by the endothelial cells of those organs

32
Q

What’s the Mechanism of Metastasis

A

Predictable Metastasis Sites: Many tumors spread to predictable sites. For instance, they often arrest in the first capillary bed they encounter, commonly the lungs or liver.
Organ Tropism: Some cancers preferentially metastasize to specific organs due to the expression of adhesion molecules or chemokine receptors that match ligands expressed by the endothelial cells of those organs.
Epithelial-Mesenchymal Transition (EMT): Genes such as TWIST and SNAIL promote EMT, a process where epithelial cells gain migratory and invasive properties, aiding in metastasis.

33
Q

Cancer cells often exhibit a metabolic shift known as the ______
Where what happens?

A

Warburg effect
where they favor glycolysis for energy production over oxidative phosphorylation, even in the presence of adequate oxygen.

34
Q

What causes the acidification of the tumor microenvironment?

A

Cancer Cells: Under the influence of growth factors, cancer cells shift towards glycolysis, even when oxygen is present. This process is less efficient in terms of ATP production but is faster and supports the high demands for energy and biosynthetic precursors needed for rapid cell growth and division.

Glucose and Lactate Production:

Glucose Utilization: Cancer cells take up large amounts of glucose and convert it into pyruvate through glycolysis.

Lactate Production: Instead of using pyruvate in the mitochondria for oxidative phosphorylation, cancer cells convert pyruvate into lactate. This happens regardless of the presence of oxygen, a phenomenon first observed by Otto Warburg in the 1920s. This results in the production of lactate and acidification of the tumor microenvironment.

35
Q

What ate the drivers for the metabolic shift in cancer cells?

A

Oncogenes:
Oncoproteins resulting from mutations in genes like RAS and MYC drive the Warburg effect. These proteins increase the expression of genes involved in glycolysis, promoting glucose uptake and conversion to lactate. This metabolic reprogramming supports the anabolic needs of proliferating cancer cells by providing building blocks for nucleotides, amino acids, and lipids.

Tumor Suppressor Genes
Loss-of-function mutations in tumor suppressor genes like PTEN, NF1, and p53 remove the inhibitory controls on glycolysis, making this metabolic shift more permanent in cancer cells.

Tumor Suppressors Opposing Warburg Effect:

Tumor suppressor genes like p53 typically oppose the Warburg effect by promoting oxidative phosphorylation and inhibiting glycolysis. Loss or mutation of these genes removes these checks and balances, facilitating the glycolytic metabolism typical of cancer cells.
For example, p53 normally activates the transcription of genes involved in oxidative phosphorylation and inhibits glycolytic enzymes. When p53 is mutated or deleted, these regulatory functions are lost, further pushing the cell towards glycolysis.

36
Q

Mechanism of Immune Evasion
Antitumor Activity occurs how

A

Cell-Mediated Immunity: The immune system primarily uses cell-mediated mechanisms to fight cancer. This involves CD8+ cytotoxic T lymphocytes (CTLs) recognizing and killing tumor cells.

MHC Class I Molecules: Tumor cells display antigens (specific protein fragments) on their surface using molecules called Major Histocompatibility Complex (MHC) class I. CTLs recognize these antigens and target the tumor cells for destruction.

37
Q

Immune Recognition and Destruction:

Tumor Cells as Nonself: Tumor cells can be recognized by the immune system as foreign (nonself) and targeted for destruction.
Cell-Mediated Mechanisms: The immune system, particularly through cell-mediated mechanisms involving CD8+ cytotoxic T lymphocytes (CTLs), plays a significant role in attacking tumor cells.
Example: Think of the immune system as a security team that identifies and eliminates intruders. In this case, the intruders are cancer cells, and the security team consists of immune cells like CTLs.
Presentation of Tumor Antigens:

MHC Class I Molecules: Tumor antigens are displayed on the surface of cancer cells by Major Histocompatibility Complex (MHC) class I molecules. These antigens are recognized by CD8+ CTLs, which then target and kill the cancer cells.
Example: Imagine MHC class I molecules as display boards showing “wanted” posters of cancer cells. CTLs act as law enforcement officers who recognize these posters and eliminate the criminals (cancer cells).
Increased Cancer Risk in Immunosuppressed Patients:

Immunosuppression and Cancer: Immunosuppressed patients, such as those undergoing organ transplants or with certain infections, have a higher risk of developing cancers, particularly those caused by oncogenic DNA viruses.
Example: Consider an immunosuppressed patient as a building with a weakened security system, making it easier for intruders (cancer cells) to invade and cause damage.

A
38
Q

What’s the Mechanisms by which tumor Immune Evasion: occur?

A

Selective Outgrowth of Antigen-Negative Variants:

Tumors can evolve to lose or reduce the expression of antigens that the immune system recognizes, allowing them to escape immune detection.

Example: Imagine cancer cells as chameleons that change their appearance to blend in and avoid being caught by the security team.

  • Loss or Reduced Expression of Histocompatibility Antigens:

Tumor cells may decrease the expression of MHC molecules, making it harder for CTLs to recognize and target them.

Example: This is akin to removing “wanted” posters from display boards, preventing law enforcement officers from identifying and capturing the criminals.

  • Immunosuppression by Tumor Cells:

Tumor cells can produce factors like TGF-β and PD-1 ligand that suppress the immune response, protecting the tumor from immune attack.

Example: Think of these factors as cloaking devices that make the cancer cells invisible to the immune system.

mmune Checkpoints and Cancer Treatment:

  • Immune Checkpoints: Tumors can exploit immune checkpoints (molecules that regulate immune responses) to inhibit immune activity. Examples include PD-1 and its ligand PD-L1, which can suppress T-cell activity.
  • Checkpoint Inhibitors:
    Antibodies that block these immune checkpoints can restore the immune system’s ability to recognize and attack cancer cells. These treatments are particularly effective against cancers with a high mutational burden.

Example: Consider checkpoint inhibitors as tools that disable the cloaking devices, making cancer cells visible to the immune system again.

39
Q

Immune Evasion Example:

Lung Cancer: Lung cancer cells often express high levels of PD-L1, which binds to the PD-1 receptor on T-cells and suppresses their activity. By using drugs that block PD-1 or PD-L1, the immune system can be reactivated to attack the cancer.

A
40
Q

Checkpoint Inhibitor Treatment Example:

Advanced Melanoma: Patients with advanced melanoma have shown significant responses to checkpoint inhibitors like pembrolizumab (Keytruda) and nivolumab (Opdivo), which block the PD-1 pathway and enhance the immune system’s ability to fight cancer.

A
41
Q

Explain How Genomic instability act as an enabler for malignancy?

A

Genomic instability refers to the increased tendency of the genome to acquire mutations, leading to genetic variations that can drive cancer development. Genetic aberrations, especially those that impair DNA repair mechanisms, play a crucial role in facilitating the acquisition of mutations that promote tumor initiation and progression.

42
Q

List How Genomic instability act as an enabler for malignancy?

A

Genetic Aberrations and Mutation Rates:

Genetic Aberrations in Cancer: Genetic changes that increase mutation rates are common in cancers. These mutations facilitate the acquisition of “driver” mutations, which are necessary for transforming normal cells into cancerous ones and promoting tumor progression.

Example: Think of genomic instability as a car with a faulty brake system, leading to frequent accidents (mutations) that can severely damage the car (cell).

Inherited DNA Repair Gene Mutations:

Increased Cancer Risk: Individuals with inherited mutations in genes responsible for DNA repair have a higher risk of developing cancer.

Example: Imagine inheriting a house with a defective security system (DNA repair genes). The house (genome) is more vulnerable to break-ins (mutations) that can lead to severe damage (cancer).

Mismatch Repair System:

Example: Consider mismatch repair as a proofreading system for a book. If the system is defective, numerous spelling errors (mutations) occur, resulting in a faulty manuscript (genome).

43
Q

List a situation where Mismatch Repair System: occur

A

HNPCC Syndrome: Hereditary nonpolyposis colorectal cancer (HNPCC) syndrome, also known as Lynch syndrome, is caused by defects in the mismatch repair system, leading to a higher risk of colon cancer. These patients exhibit microsatellite instability, characterized by changes in the length of short DNA repeats throughout the genome.

Example: Consider mismatch repair as a proofreading system for a book. If the system is defective, numerous spelling errors (mutations) occur, resulting in a faulty manuscript (genome).

Xeroderma Pigmentosum

Homologous Recombination DNA Repair:

44
Q

What’s (Nucleotide Excision Repair) Xeroderma Pigmentosum:

A

Xeroderma Pigmentosum: Patients with xeroderma pigmentosum have a defect in the nucleotide excision repair pathway, making them highly susceptible to skin cancers caused by UV light because they cannot repair pyrimidine dimers effectively.

Example: Think of nucleotide excision repair as a sunscreen protecting against UV damage. Without it, the skin is highly vulnerable to damage (mutations) and cancer.

45
Q

Disorders with Homologous Recombination Defects can cause what type of dxs

A

Homologous Recombination DNA Repair:

Disorders with Homologous Recombination Defects:
Conditions like Bloom syndrome, ataxia-telangiectasia, and Fanconi anemia involve defects in the homologous recombination DNA repair system. These disorders are characterized by developmental issues and increased sensitivity to DNA-damaging agents like ionizing radiation.

Example: Homologous recombination repair is like a repair crew fixing a damaged bridge (DNA). If the crew is ineffective, the bridge remains unstable and prone to collapse (mutations).

46
Q

Mutations in what genes increases risk of breast cancer?

A

Breast Cancer Risk: Mutations in BRCA1 and BRCA2 genes, which are involved in DNA repair, significantly increase the risk of developing familial breast cancers.

Example: BRCA genes are like specialized mechanics for repairing a complex machine (DNA). If they are missing or defective, the machine is more likely to break down (develop cancer).

47
Q

Mutations in what systems can cause or increase risk of cancer?

A

BRCA1 and BRCA2 genes = Breast

Regulated Genomic Instability in T and B Cells:

Proofreading Function of DNA Polymerase:

Disorders with Homologous Recombination Defects:

Nucleotide Excision Repair:

48
Q

_____ and ______ play a significant role in promoting various hallmarks of cancer, such as proliferation, invasion, and metastasis

A

Inflammatory cells & the local microenvironment

49
Q

Chronic Inflammatory Reaction:

Inflammation and Cancer: Infiltrating cancers provoke a chronic inflammatory response. This inflammation modifies tumor cells and their surrounding environment to enable cancer progression.
Example: Think of cancer as a wound that keeps getting worse instead of healing. The continuous inflammatory response to this “wound” fuels cancer growth

A
50
Q

Inflammatory Cells and Microenvironment:

Modification of Tumor Cells: Inflammatory cells, including macrophages, mast cells, neutrophils, and T and B lymphocytes, interact with tumor cells and the local microenvironment. These interactions promote cancer progression.

Example: Imagine a construction site where workers (inflammatory cells) not only fix issues but also inadvertently help expand the building (cancer).

A
51
Q

Cancer-Enabling Effects of Inflammatory and Stromal Cells:

Release of Proliferative Factors: Inflammatory and stromal cells release factors such as ………..
These factors promote tumor cell proliferation, tissue invasion, and metastasis.

Example: Think of these factors as fertilizers that not only help plants (cancer cells) grow but also enable them to spread to new areas (metastasize).

A

Epidermal growth factor (EGF),
Vascular endothelial growth factor (VEGF-A/C)
Fibroblast growth factor 2 (FGF2),
Chemokines,
Cytokines,
Matrix metalloproteinase 9 (MMP-9), and
Cysteine cathepsin proteases.