Chapter 20 Omega

0.0(0)
studied byStudied by 0 people
0.0(0)
full-widthCall Kai
learnLearn
examPractice Test
spaced repetitionSpaced Repetition
heart puzzleMatch
flashcardsFlashcards
GameKnowt Play
Card Sorting

1/84

encourage image

There's no tags or description

Looks like no tags are added yet.

Study Analytics
Name
Mastery
Learn
Test
Matching
Spaced

No study sessions yet.

85 Terms

1
New cards

Cancer

A disease in which a clone of cells reproduces despite normal restraints and invades tissues.

2
New cards

Benign

A tumor that grows locally but does not invade surrounding tissue.

3
New cards

Malignant

A tumor that invades nearby tissue and can spread to distant sites.

4
New cards

Metastasis

The spread of cancer cells through blood or lymph to form secondary tumors.

5
New cards

Carcinoma

A cancer arising from epithelial cells.

6
New cards

Sarcoma

A cancer arising from connective tissue or muscle cells.

7
New cards

Leukemia

A cancer of blood-forming cells often producing abnormal white blood cells.

8
New cards

Lymphoma

A cancer originating from lymphoid (immune) cells.

9
New cards

Adenoma

A benign epithelial tumor with glandular organization.

10
New cards

Tumor (neoplasm)

A growth produced by excessive cell proliferation

11
New cards

Cancer as microevolution

Cancer develops through mutation, competition, and natural selection among somatic cells.

12
New cards

Clonal origin of cancer

Most cancers arise from a single abnormal cell that expands into a clone.

13
New cards

Somatic mutation

A DNA change in body cells passed to daughter cells that can contribute to cancer.

14
New cards

Carcinogenesis external agents

Cancer can result from chemical carcinogens or radiation that alter DNA.

15
New cards

Why cancer is relatively infrequent

Cancer requires multiple rare genetic changes, usually taking decades to accumulate.

16
New cards

Tumor progression timing

Cancers develop slowly because many sequential mutations are needed.

17
New cards

Natural selection in tumors

Mutant cells with growth advantages outcompete others, accelerating tumor evolution.

18
New cards

Genetic instability

Cancer cells often have high mutation rates and chromosomal abnormalities.

19
New cards

Aneuploidy

Gain or loss of whole chromosomes, common in cancer cells.

20
New cards

Chromothripsis

A catastrophic chromosome shattering and reassembly event causing massive genomic changes.

21
New cards

Cancer stem cells

A subpopulation of tumor cells capable of self-renewal and regenerating the tumor.

22
New cards

Six common hallmarks of cancer

Altered homeostasis, bypassing proliferation limits, evading apoptosis, altered metabolism, manipulating tissue/immune environment, and metastasis.

23
New cards

Altered homeostasis

Cancer cells divide faster than they die or avoid apoptosis, causing net growth.

24
New cards

Contact inhibition

Normal cells stop dividing when contacting neighbors

25
New cards

Replicative senescence

A limit to cell divisions due to telomere shortening

26
New cards

How tumor cells avoid senescence

They reactivate telomerase or use ALT to maintain telomeres.

27
New cards

Apoptosis evasion vs necrosis

Cancer cells block apoptosis but inner tumor cells often die from necrosis

28
New cards

Warburg effect

Cancer cells rely heavily on glycolysis and glucose uptake even with oxygen present.

29
New cards

Tumor stroma

Supportive connective tissue and cells surrounding the tumor that help its growth.

30
New cards

Tumor–stroma co-evolution

Cancer and stromal cells influence each other, promoting tumor survival and angiogenesis.

31
New cards

Angiogenesis

The formation of new blood vessels to supply the growing tumor.

32
New cards

Metastasis steps

Invasion, intravasation, circulation survival, extravasation, and colonization at a new site.

33
New cards

Why metastasis is rare

Few circulating tumor cells survive and successfully colonize distant tissues.

34
New cards

Oncogenes vs tumor suppressors

Oncogenes stimulate growth when activated

35
New cards

Retroviruses and oncogenes

Retroviruses can activate or introduce oncogenes that drive cancer.

36
New cards

Ras in cancer

Ras becomes oncogenic when mutations lock it in an active state, driving proliferation.

37
New cards

Ways proto-oncogene → oncogene

Point mutation, regulatory mutation, gene amplification, or chromosomal rearrangement.

38
New cards

Retinoblastoma (Rb) genetics

Loss of both Rb copies causes tumors

39
New cards

Ways to lose tumor-suppressor copy

Deletion, mutation, recombination, chromosome loss, or epigenetic silencing.

40
New cards

Cancer-critical genes

Genes whose mutation drives cancer development.

41
New cards

PI3K/Akt/mTOR pathway

A growth-promoting pathway often overactive in cancer.

42
New cards

PTEN

A phosphatase that inhibits PI3K signaling

43
New cards

p53 pathway

p53 activates repair, arrest, or apoptosis

44
New cards

Drivers vs passengers

Drivers directly promote cancer

45
New cards

Oncogene dependence

Some cancers rely on one key oncogene and respond to targeted therapy against it.

46
New cards

Cancer genome projects

Large sequencing efforts identify common mutations and drivers in cancers.

47
New cards

Epigenetic contributions

DNA methylation and histone changes can silence or activate cancer-related genes.

48
New cards

Aneuploidy prevalence

Most human cancers display characteristic chromosome-number abnormalities.

49
New cards
Drivers vs passengers
Driver mutations causally promote cancer growth and are recurrent across tumors, while passenger mutations are incidental byproducts of genomic instability
50
New cards
PI3K/Akt/mTOR pathway mutations
Activating mutations or loss of negative regulators (e.g., PTEN loss) hyperactivate PI3K→Akt→mTOR signaling, driving anabolic growth, increased glucose uptake and lipid/protein synthesis, and promoting tumor growth and the Warburg metabolic phenotype.
51
New cards
p53 activation and cancer
p53 is raised by DNA damage, telomere loss, hypoxia, oxidative/osmotic stress, and excessive oncogenic signaling (e.g., Myc)
52
New cards
Colorectal cancer — physiological progression
Normal epithelium → benign adenomatous polyps (adenomas) → accumulation of mutations and dysplasia → invasive carcinoma → possible metastasis
53
New cards
FAP (familial adenomatous polyposis) genetics
FAP is caused by inherited loss-of-function mutations in one APC allele (germline), leading to early, numerous colorectal polyps that acquire a second hit in APC in colon cells, driving near-certain progression to cancer if untreated.
54
New cards
HNPCC / Lynch syndrome genetics
HNPCC (Lynch) is due to inherited defects in DNA mismatch-repair genes (e.g., MLH1, MSH2), which greatly increase mutation rates when the remaining allele is lost, predisposing to colorectal cancer without massive polyposis and producing microsatellite instability.
55
New cards
Sequence of genetic changes in colorectal cancer
Commonly: early APC loss → expansion of abnormal crypt/stem cells → later activation of K-Ras in enlarging polyps → subsequent loss of p53 and other alterations → progression from adenoma to carcinoma (with variability across cases).
56
New cards
Small-molecule therapy for CML (Bcr-Abl inhibition)
CML is driven by the Bcr-Abl fusion tyrosine kinase (Philadelphia chromosome)
57
New cards
Directing the immune system to cancer
Strategies include monoclonal antibodies (blocking or toxin-conjugated), adoptive T-cell transfer/expansion, CAR-T cell engineering, and vaccines—these increase tumor antigen recognition and T-cell–mediated killing of cancer cells.
58
New cards
Removing tumor immunosuppression (checkpoint blockade) and risks
Checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4) or blockade of tumor immunosuppressive signals can unleash T cells to kill tumors but may cause systemic immune hyperactivation and autoimmunity (severe inflammatory side effects).
59
New cards
Cancer stem cells
Some tumors contain a rare subpopulation of self-renewing cancer stem cells that generate most tumor cells (transit-amplifying progeny). They are often slowly dividing and can repopulate the tumor after treatment, making cure difficult because therapies that kill rapidly dividing cells may spare these stem cells.
60
New cards
Six hallmarks of cancer
The common hallmarks are: (1) altered homeostasis (growth > death), (2) bypass of proliferation limits, (3) evasion of cell-death signals, (4) altered metabolism, (5) manipulation of the tissue/immune environment, and (6) escape and metastasis.
61
New cards
Altered homeostasis (apoptosis vs division)
Cancer arises when cell division outpaces cell death—either by increased proliferation or by evading apoptosis—causing net tumor growth.
62
New cards
Contact inhibition
A normal process where cells stop dividing/moving when they touch neighbors
63
New cards
Replicative cell senescence
A permanent arrest of cell division after many population doublings (telomere shortening triggers DNA-damage signals), serving as a barrier to unlimited proliferation.
64
New cards
Why cells undergo senescence
Telomere erosion and resulting DNA-damage signals activate cell-cycle arrest to prevent genomic instability and malignant transformation.
65
New cards
How cancer cells avoid senescence
Most reactivate telomerase to maintain telomeres
66
New cards
Do tumor cells undergo apoptosis
Tumor cells commonly disable apoptosis pathways to survive stress, but many cells within large tumors still die by necrosis due to hypoxia and nutrient shortage
67
New cards
Warburg effect
Tumor cells take up large amounts of glucose and favor aerobic glycolysis (producing building blocks for growth) even when oxygen is present, supporting rapid proliferation and enabling glucose-based imaging.
68
New cards
Tumor stroma
The supportive connective tissue around a tumor (fibroblasts, immune cells, endothelial cells, extracellular matrix) that provides structural support and signals the cancer needs to grow.
69
New cards
Tumor–stroma co-evolution
Cancer cells secrete signals and proteases that remodel stroma
70
New cards
Angiogenesis in tumors
Tumors induce formation of new blood vessels to supply oxygen and nutrients as they grow, often via stromal signaling.
71
New cards
Process of metastasis
Steps: local invasion → intravasation into lymph/blood → survival in circulation → extravasation into distant tissue → colonization and proliferation to form a secondary tumor.
72
New cards
Why metastasis is rare
Although many cells enter circulation, very few survive, adapt to foreign microenvironments, and successfully colonize—colonization is the rate-limiting, low-probability step.
73
New cards
Oncogenes vs tumor suppressors
Oncogenes result from gain-of-function changes (act dominantly) that promote growth
74
New cards
Gain-of-function vs loss-of-function
Gain-of-function mutations (oncogenes) typically require only one altered allele to drive cancer
75
New cards
How retroviruses cause cancer
Retroviruses can insert into the host genome, activate nearby cellular proto-oncogenes, or carry captured viral oncogenes (v-onc) that are mutated versions of host proto-oncogenes, driving transformation.
76
New cards
Ras in cancer
Ras proteins are GTPases that relay growth signals
77
New cards
Ways proto-oncogene → oncogene
Point or regulatory mutations, gene amplification (extra copies), chromosomal rearrangements that create fusion proteins or place genes under strong promoters, and deletions that produce constitutive activity.
78
New cards
Example of receptor activation
Deletion or mutation of extracellular domains (eg. in some EGF receptor mutants) can make receptors constitutively active without ligand, driving proliferation.
79
New cards
Retinoblastoma genetics
Retinoblastoma arises from loss of Rb tumor-suppressor function (a loss-of-function mutation). Hereditary cases inherit one defective Rb allele and often develop bilateral/multiple tumors
80
New cards
Two-hit model for tumor suppressors
Tumor suppressor inactivation typically follows two events: one allele lost/inactivated first (sometimes inherited) and the second allele lost later somatically, eliminating gene function.
81
New cards
Ways to lose the wildtype tumor-suppressor copy
Small deletions, point mutations, mitotic recombination/gene conversion, chromosome loss, large rearrangements, or epigenetic silencing (promoter methylation).
82
New cards
Genetic vs epigenetic inactivation
Genetic changes alter DNA sequence (deletions, mutations, LOH)
83
New cards
Cancer-critical genes
Genes whose alteration drives cancer progression (drivers) as opposed to incidental passenger mutations
84
New cards
Genome disruption in cancer
Many cancers show gross genomic disorder—point mutations, copy-number changes, chromosomal rearrangements, aneuploidy, and catastrophic events like chromothripsis—contributing to heterogeneity.
85
New cards
Epigenetic contributions to cancer
Aberrant DNA methylation and histone modifications can silence tumor suppressors or alter gene networks