Neoplasia

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

.

1, Neoplasia:

• Definition: Uncontrolled new growth of cells that forms a mass (tumor).


These tumors can be benign or malignant.
• Key Points:
o Disruption of normal cell proliferation and differentiation pathways.
o Can arise from any cell type in the body.
o Not synonymous with cancer (cancer is a subset of neoplasia).

2. Benign Neoplasms:

• Definition: Slow-growing, localized tumors that do not invade or spread.


• Characteristics:
o Well-defined capsule (often).
o Well-differentiated cells with morphology and function resembling
normal cells.
o Low mitotic activity (cell division).
o Generally good prognosis.

Examples:

• Mesenchymal Origin:
o Lipoma: Soft, doughy tumors arising from adipose tissue (fat cells).
o Leiomyoma: Firm tumors derived from smooth muscle cells (e.g.,
uterus).
• Epithelial Origin:
o Papilloma: Wart-like growths composed of squamous epithelial cells
(e.g., skin).
o Adenoma: Polypoid tumors originating from glandular epithelium (e.g.,
colon). May secrete hormones depending on cell type.

3. Malignant Neoplasms (Cancer):

• Definition: Aggressive tumors with the potential to invade locally, spread to


distant organs (metastasis), and cause significant morbidity and mortality.
• Characteristics:
o Uncontrolled growth.
o Local invasion and destruction of surrounding tissues.
o Metastasis via lymphatic or blood vessels.
o Anaplastic cells with abnormal morphology (loss of differentiation).
o Increased mitotic activity.
o Ability to evade cell death (apoptosis).
o Variable prognosis depending on tumor type, stage, and treatment.

Examples:

• Mesenchymal Origin:
o Sarcoma: Malignant tumor arising from connective tissues (e.g.,
osteosarcoma from bone).
o Fibrosarcoma: Cancerous tumor derived from fibroblasts (fibrous
connective tissue).
• Epithelial Origin:
o Carcinoma: Malignant tumor originating from epithelial tissue. Specific
types named based on tissue of origin (e.g., lung carcinoma, breast
adenocarcinoma).

4. Mixed Tumor:

• Uncommon neoplasm composed of multiple cell types from different tissues.


• Can be benign (e.g., pleomorphic adenoma of salivary gland with epithelial
and mesenchymal elements) or malignant.

5. Teratoma:

• Specific type of germ cell tumor containing tissues derived from all three
embryonic germ layers (endoderm, mesoderm, ectoderm).
• Tissues can be mature or immature and arranged in a disorganized manner.
• Can be benign or malignant (benign more common).

6. Distinguishing Benign vs. Malignant Neoplasms:

Feature Benign Neoplasm Malignant Neoplasm (Cancer)

Growth Rate Slow Fast

Local Invasion No Yes (invasive)

Metastasis No Yes (spreads to distant sites)

Cell Differentiation Well-differentiated Poorly differentiated (anaplastic)


(similar to normal cells)

Capsule Often present Usually absent


Presence

Prognosis Generally good Variable, can be life-threatening

Cytomorphology Cells resemble normal, Pleomorphism, hyperchromasia, increased


low mitotic activity nuclear-to-cytoplasmic ratio, abnormal
mitoses

7. Cytomorphological Differences:
• Benign: Cells with normal size, shape, and nuclear features. Low mitotic
activity.
• Malignant: Pleomorphic cells (variation in size and shape), hyperchromatic
nuclei (darkly stained), increased nuclear-to-cytoplasmic ratio, and abnormal
mitotic figures.

8. Anaplasia:

• Loss of differentiation in cancer cells.


• Cells lose their resemblance to their tissue of origin and appear abnormal in
size, shape, and nuclear features.
• Prominent nucleoli and abnormal mitotic figures are often seen.

9. Dysplasia:

• Precancerous condition with abnormal cellular proliferation and maturation


within an epithelium.
• Cells show some architectural disorganization but remain confined to the
epithelium (no invasion).
• Can be a warning sign for potential future malignancy.

10. Carcinoma in situ:

• Specific type of dysplasia involving the epithelial layer.


• Abnormal cells are confined to the basement membrane and haven't invaded
the underlying

11. Metastasis:

• The process by which cancer cells detach from the primary tumor, travel
through the bloodstream or lymphatic system, and establish secondary
tumors in distant organs.

12. Pathways of Spread in Metastasis:

• Lymphatic Spread:
o Cancer cells enter lymphatic vessels and travel to regional lymph
nodes, which can become involved in the tumor process.
o Example: Breast cancer cells often metastasize to axillary lymph nodes
first.
• Hematogenous Spread:
o Cancer cells gain access to the bloodstream and can be carried
throughout the body, potentially seeding distant organs.
o Example: Lung cancer cells can metastasize to the brain or bones via
hematogenous spread.

13. Occupational Cancer (Short Note):


• Exposure to certain workplace hazards can increase the risk of developing
specific cancers.
• Examples:
o Asbestos exposure: Linked to mesothelioma (lung cancer) and lung
cancer.
o Benzene exposure: Associated with increased risk of leukemia.
o Ultraviolet radiation: A major risk factor for skin cancer.

14. Chronic Inflammation and Cancer:

• Chronic inflammation can increase cancer risk:


o Inflammatory processes can damage DNA and create a
microenvironment that promotes cell proliferation.
o Inflammatory cells can release growth factors and signaling molecules
that can contribute to tumor development.
o Example: Chronic ulcerative colitis (inflammatory bowel disease) is a
risk factor for colon cancer.
• Infiltrating cancer can provoke chronic inflammation:
o Cancer cells can release inflammatory mediators, attracting immune
cells to the tumor microenvironment.
o This chronic inflammatory response can contribute to tumor growth,
angiogenesis (new blood vessel formation), and metastasis.

15. Eight Hallmarks of Cancer (Hanahan and Weinberg):

1. Sustaining proliferative signaling: Cancer cells acquire the ability to


continuously divide and grow independent of external signals.
2. Evading growth suppressors: Mutations inactivate tumor suppressor genes
that normally restrain cell proliferation.
3. Resisting cell death (apoptosis): Cancer cells develop mechanisms to
evade programmed cell death pathways.
4. Enabling replicative immortality: Cancer cells bypass the Hayflick limit
(finite cell divisions) and achieve limitless replication potential.
5. Inducing angiogenesis: Tumors promote the growth of new blood vessels to
supply nutrients and oxygen for their expansion.
6. Activating invasion and metastasis: Cancer cells acquire the ability to
invade surrounding tissues, enter the lymphatic or blood vessels, and
establish secondary tumors.
7. Promoting tumor-promoting inflammation: Tumors create an inflammatory
microenvironment that fosters growth, survival, and metastasis.
8. Deregulating cellular metabolism: Cancer cells reprogram their metabolism
to favor energy production and biosynthesis for rapid growth, often relying on
aerobic glycolysis (Warburg effect).

16. Genes Targeted by Cancer-Causing Mutations:

• Cancer arises from a multistep process involving the accumulation of


mutations in various genes.
• These genes can be broadly categorized into two main groups:
o Oncogenes: Genes that promote cell growth and proliferation when
mutated.
o Tumor suppressor genes: Genes that normally restrain cell
proliferation and, when mutated or inactivated, contribute to cancer
development.

17. Driver Mutations:

• Driver mutations are the key genetic alterations that provide a growth
advantage to cells and propel them towards malignancy.
• These mutations can occur in oncogenes or tumor suppressor genes.

18. Oncogenes:

• Genes with the potential to transform normal cells into cancer cells when
mutated or activated.
• Examples of Proto-oncogenes and their Activation:
o RAS proto-oncogenes: Frequently mutated in various cancers (e.g.,
colon, lung). Mutations lead to constitutive activation of signaling
pathways that promote cell proliferation.
o MYC proto-oncogene: Encodes a transcription factor involved in cell
growth and proliferation. Amplification or overexpression of MYC is
seen in many cancers (e.g., breast, lung).

19. Tumor Suppressor Genes:

• Genes that normally inhibit cell proliferation and promote DNA repair.
• Mutations or deletions in tumor suppressor genes can contribute to
uncontrolled cell growth and cancer development.
• Examples with Familial Syndromes and Sporadic Cancers:
o TP53 (p53) gene: "Guardian of the genome." Mutations in TP53 are
found in a wide range of cancers. Hereditary mutations predispose to
Li-Fraumeni syndrome (multiple cancers).
o RB1 (retinoblastoma) gene: "Governor of proliferation." Controls cell
cycle progression. Mutations cause retinoblastoma (eye cancer) in
children and can contribute to other cancers.

20. RB Gene (Retinoblastoma Gene) - The Governor of Proliferation

• Function: The RB gene encodes a protein called retinoblastoma protein


(pRB). pRB acts as a tumor suppressor by regulating cell cycle progression.
• Mechanism: pRB binds to proteins like E2F transcription factors, preventing
them from activating genes required for S phase (DNA replication) entry. This
keeps cell proliferation in check.
• Mutations: Inactivation of the RB gene through mutations allows uncontrolled
E2F activity, leading to unchecked cell cycle progression and tumor formation.
• Clinical significance: Mutations in RB cause retinoblastoma, a childhood
eye cancer. Rb gene alterations are also implicated in various other cancers,
including osteosarcoma, lung cancer, and breast cancer. (Robbins & Cotran,
Pathologic Basis of Disease, Chapter 10)

21. TP53 Gene (Tumor Protein 53) - The Guardian of the Genome

• Function: The TP53 gene encodes a protein called p53, a critical tumor
suppressor that plays a multifaceted role in maintaining genomic integrity.
• Mechanisms: p53 has various functions:
o Cell Cycle Arrest: Upon DNA damage, p53 activates genes that halt
cell cycle progression, allowing time for DNA repair.
o DNA Repair: p53 upregulates genes involved in DNA repair
mechanisms.
o Apoptosis: If DNA damage is severe and unrepairable, p53 triggers
apoptosis (programmed cell death) to eliminate potentially dangerous
cells.
• Mutations: Mutations in the TP53 gene are the most frequent genetic
alterations observed in human cancers. Loss of functional p53 prevents these
critical functions, leading to uncontrolled cell proliferation and tumorigenesis.
• Clinical significance: TP53 mutations are found in a wide range of cancers,
including colon cancer, lung cancer, breast cancer, and many others.
(Robbins & Cotran, Pathologic Basis ofDisease, Chapter 10)

22. APC Gene and Regulation of β-catenin

• Function: The APC (Adenomatous Polyposis Coli) gene encodes a protein


involved in the Wnt signaling pathway, essential for embryonic development
and adult tissue homeostasis.
• β-catenin Regulation: APC acts as a negative regulator of β-catenin, a
protein that functions as a transcriptional co-activator. In the absence of Wnt
signaling, APC targets β-catenin for degradation.
• Wnt Signaling Activation: When Wnt signaling is activated, APC
phosphorylation is inhibited, leading to β-catenin stabilization and
translocation to the nucleus. There, β-catenin interacts with TCF/LEF
transcription factors, promoting the expression of genes involved in cell
proliferation, differentiation, and migration.
• APC Mutations and Tumorigenesis: Mutations in the APC gene lead to
constitutive activation of Wnt signaling due to β-catenin stabilization. This
uncontrolled proliferation and migration contribute to tumor development.
• Clinical significance: APC mutations are a hallmark of Familial
Adenomatous Polyposis (FAP), a hereditary condition predisposing
individuals to colon cancer. APC mutations are also found in sporadic
colorectal cancers. (Robbins & Cotran, Pathologic Basis of Disease, Chapter
14)

23. Warburg Effect

• Observations: In the early 1920s, Otto Warburg observed that cancer cells,
even in the presence of oxygen, preferentially rely on aerobic glycolysis for
energy production (fermentation) instead of mitochondrial oxidative
phosphorylation, which is more efficient.
• Causes: The reasons for the Warburg effect are complex and not fully
understood. Factors like mitochondrial dysfunction, altered gene expression
patterns, and the need for rapid ATP production to support cell proliferation
might contribute.
• Consequences: The Warburg effect has several consequences for cancer
cells:
o Increased production of lactic acid, leading to a more acidic tumor
microenvironment.
o Altered metabolism promotes the synthesis of building blocks needed
for rapid cell growth.
o The Warburg effect is a potential target for cancer therapies. (Robbins
& Cotran, Pathologic Basis of Disease, Chapter 10)

24. Oncometabolism

• Definition: Oncometabolism refers to the altered metabolic state of cancer


cells characterized by the Warburg effect and other metabolic changes.
• Features: Oncometabolism encompasses a spectrum of metabolic
reprogramming events in cancer cells:
o Increased glucose uptake and reliance on aerobic glycolysis.
o Altered amino acid metabolism to support rapid protein synthesis.
o Fatty acid metabolism reprogramming for energy and membrane
synthesis.
o Changes in mitochondrial function.
• Significance: Understanding oncometabolism provides insights into cancer
cell vulnerabilities and potential therapeutic targets. By manipulating cancer
cell metabolism

25. Mechanisms of Apoptosis Evasion in Cancer

Cancer cells employ various mechanisms to bypass or disable the apoptotic


machinery:

1. Mutations in tumor suppressor genes: Mutations in genes like TP53 (p53)


can impair the apoptotic response.
2. Deregulation of apoptotic signaling pathways: Alterations in signaling
pathways that normally trigger apoptosis can occur.
3. Inactivation of caspases: Cancer cells can produce caspase inhibitors or
develop caspase deficiencies.
4. Enhanced DNA repair mechanisms: Increased DNA repair activity allows
cancer cells to avoid p53-mediated apoptosis.
5. Alterations in the tumor microenvironment: The tumor microenvironment
can promote survival signals and inhibit apoptosis.

26. The Metastatic Cascade

The metastatic cascade is a multistep process by which cancer cells spread from the
primary tumor to distant organs:
1. Local Invasion: Cancer cells lose cell-adhesion properties and degrade the
extracellular matrix.
pen_spark

2. Intravasation: Cancer cells enter lymphatic or blood vessels.


3. Dissemination: CTCs travel through the bloodstream or lymphatic system.
4. Extravasation: CTCs exit the bloodstream or lymphatic vessels and migrate
into distant organs.
5. Growth at the Secondary Site: CTCs proliferate at the new location, forming
a metastatic tumor.

Opens in a new window www.researchgate.net


Metastatic Cascade

27. Tumor Antigens

Tumor antigens are molecules that the immune system recognizes as foreign in the
context of cancer. These antigens can be:

1. Tumor-associated antigens (TAAs): These are self-antigens that are


normally expressed in low amounts or in restricted cell types but are
overexpressed or aberrantly expressed in cancer cells.
2. Tumor-specific antigens (TSAs): These are antigens arising from mutations
or viral integrations specific to cancer cells.

28. Viral and Bacterial Oncogenesis

Viruses:

• Mechanism: Oncogenic viruses encode viral genes that can disrupt various
cellular processes leading to cancer development. Key mechanisms include:
o Disruption of cell cycle control: Viral proteins can interact with and
inactivate tumor suppressor proteins like pRB, leading to uncontrolled
cell proliferation.
o Activation of growth signaling pathways: Viruses can encode
oncogenes that mimic growth factors or activate signaling pathways
like Ras-ERK, promoting cell proliferation and survival.
o Inhibition of apoptosis: Viral proteins can interfere with apoptotic
signaling pathways, allowing infected cells to evade cell death.
Genomic instability: Viral integration into the host genome can disrupt
o
genes and cause chromosomal abnormalities, increasing the risk of
mutations.
• Examples:
o Human Papillomavirus (HPV): A well-established cause of cervical
cancer and some head and neck cancers. HPV encodes proteins that
disrupt the function of pRB and p53, promoting uncontrolled cell
division.
o Hepatitis B Virus (HBV) and Hepatitis C Virus (HCV): Chronic
infection with these viruses can lead to liver cancer. Viral proteins can
induce chronic inflammation and DNA damage, promoting mutations in
hepatocytes.
o Epstein-Barr Virus (EBV): Associated with Burkitt's lymphoma and
nasopharyngeal carcinoma. EBV can immortalize B lymphocytes and
disrupt cell cycle regulation.
o Human T-lymphotropic Virus Type 1 (HTLV-1): Causes Adult T-cell
leukemia/lymphoma (ATL). HTLV-1 integrates into the genome of T
cells, promoting their proliferation and survival.

Bacteria:

• Mechanism: While less common than viral oncogenesis, chronic bacterial


infections can contribute to cancer development through several mechanisms:
o Chronic inflammation: Persistent bacterial infection can induce
chronic inflammation. Inflammatory mediators can damage host cells
and promote cell proliferation, increasing the risk of mutations.
o Production of genotoxins: Some bacteria can produce toxins that
damage DNA, leading to mutations.
o Disruption of cell cycle regulation: Certain bacterial products can
interfere with cell cycle checkpoints, promoting uncontrolled cell
division.
• Examples:
o Helicobacter pylori: Infection with this bacterium is strongly
associated with an increased risk of stomach cancer. H. pylori infection
can induce chronic inflammation and disrupt cell cycle regulation in
gastric epithelial cells.
o Salmonella spp.: Chronic Salmonella infection has been linked to an
increased risk of colon cancer, possibly due to chronic inflammation
and DNA damage.

Importance:

Understanding the mechanisms of viral and bacterial oncogenesis is crucial for:

• Developing preventive strategies: Vaccination against oncogenic viruses


like HPV and HBV can significantly reduce cancer burden.
• Identifying individuals at high risk: Screening individuals with chronic
infections associated with cancer risk allows for early detection and
intervention.
• Development of therapeutic approaches: Targeting viral or bacterial factors
involved in cancer development can be a potential therapeutic strategy.

Note: This explanation provides a deeper dive into viral and bacterial oncogenesis
but remains within the scope of a pathology student's understanding. For more
advanced details, you might consult specialized microbiology or virology texts.

29. Examples of Carcinogenic Agents and Associated Cancers:

Carcinogen Examples of Cancers


Chemical Carcinogens
- Alkylating agents Lung cancer, bladder cancer, leukemia

- Aromatic hydrocarbons Lung cancer, skin cancer


- Nitrosamines Stomach cancer, esophageal cancer
- Asbestos Mesothelioma, lung cancer
- Vinyl chloride Liver cancer, angiosarcoma
Radiation Skin cancer, leukemia, bone cancer
Ultraviolet Radiation (UV) Skin cancer

Hereditary Factors Breast cancer, colon cancer, melanoma

Lifestyle Factors
- Tobacco smoking Lung cancer, head and neck cancer,
bladder cancer, esophageal cancer
- Alcohol consumption Liver cancer, esophageal cancer,
head and neck cancer
- Diet and obesity Colon cancer, pancreatic cancer,
endometrial cancer
Infectious Agents
- Viruses (e.g., HPV, HBV) Cervical cancer, liver cancer

- Bacteria (e.g., H. pylori) Stomach cancer

30. Cancer Cachexia:

• A complex metabolic syndrome characterized by:


o Progressive weight loss (muscle wasting)
o Fatigue
o Anorexia (loss of appetite)
o Anemia
o Systemic inflammation
• Mechanisms: Not fully understood, but likely involve a combination of factors:
o Tumor-derived factors like cytokines and inflammatory mediators that
alter metabolism.
o Increased energy expenditure due to tumor growth.
o Altered protein synthesis and breakdown.
o Resistance to anabolic hormones (insulin).
• Consequences: Cachexia significantly reduces quality of life and can worsen
cancer prognosis.

Paraneoplastic Syndromes (Robbins & Cotran,


Chapter 10)
31. Paraneoplastic Syndromes:

• A group of clinical signs and symptoms caused by a tumor but not directly
related to local tumor invasion or metastasis.
• Mechanisms:
o Ectopic hormone production by tumor cells.
o Immune response to tumor antigens.
o Release of substances that affect distant organs.
• Examples:
o Hypercalcemia (increased blood calcium) associated with some lung
and breast cancers.
o Cushing's syndrome (symptoms due to excess cortisol) associated with
lung carcinoid tumors.
o Lambert-Eaton myasthenic syndrome (neuromuscular weakness)
associated with small cell lung cancer.
• Importance: Paraneoplastic syndromes can be the presenting feature of
cancer and can help guide diagnosis.

Laboratory Diagnosis of Cancer (Robbins & Cotran,


Chapter 14)
32. Laboratory Diagnosis of Cancer:

• No single test definitively diagnoses cancer. Diagnosis typically involves a


combination of approaches:
o Clinical history and physical examination: Identifying risk factors
and suspicious symptoms.
o Imaging studies: X-rays, CT scans, MRIs to visualize tumors.
o Biopsy: Obtaining a tissue sample for microscopic examination by a
pathologist.
o Cytology: Microscopic examination of cells from body fluids or
aspirates (e.g., Pap smears).
• Additional tests: Depending on the suspected cancer type, additional tests
like:
o Immunohistochemistry: Detecting specific proteins in tumor cells
using antibodies.
o Molecular testing: Analyzing mutations or genetic alterations in tumor
cells.

Tumor Markers (Robbins & Cotran, Chapter 14)


33. Tumor Markers:

• Substances elevated in the blood or other body fluids of some cancer


patients.
• Not specific for cancer and can be elevated in other conditions.
• Examples:
o Prostate-specific antigen (PSA): Elevated PSA may indicate prostate
cancer, but also prostatitis or benign enlargement.
o Carcinoembryonic antigen (CEA): Elevated CEA may suggest colon
cancer, but can also be elevated in other cancers and non-malignant
conditions.
o CA 125: Elevated CA 125 may indicate ovarian cancer, but can also be
elevated in benign conditions

You might also like